PHYLOGENETICS OF EFFECTOR MOLECULES INVOLVED IN CELL-MEDIATED CYTOTOXICITY by KESAVANNAIR PRAVEEN (Under the Direction of Liliana Jaso-Friedmann) ABSTRACT Cytotoxic lymphocytes induce target cell death using common effector pathways that involve either the components of their cytotoxic granules or members of TNF superfamily ligands. We present the first evidence for the existence of both granule exocytosis as well as TNF family ligand mediated killing in cytotoxic cells from ectothermic vertebrates. Nonspecific cytotoxic cells (NCC) are the first identified killer cell population in teleosts. This study presents the molecular evidence for expression of multiple granzymes with different substrate specificities as well as other components of cytotoxic granules in NCC, providing the sequence information for such molecules for the first time in a non-mammalian species. The main substrate specificities for granzymes found in these cells were chymase and tryptase. Teleost granzymes conserve the genomic organization described for mammalian granzymes. Teleost granzymes with chymase activity have novel S1 specificity triplet residues. This study also demonstrates the role of TNFalpha in NCC functions as an effector molecule of cytotoxicity and in regulation of NCC functions. Teleost cytotoxic cells express both membrane-bound and secreted forms of TNFalpha, which can induce cell death in susceptible target cells. Membrane-bound TNF-alpha on NCC can mediate activation-induced cell death in other NCCs in a paracrine manner, while the soluble TNF-alpha can inhibit this and protect the killer cells from activation-induced cell death. Soluble TNF-alpha upregulates the expression of other cytotoxic effector molecules like granzymes in teleost killer cells, indicating a major role played by this molecule in NCC functions. TNF-alpha-induced protection of NCC was shown to be mediated though different regulators of apoptosis. One of the important mediators of such function was identified as cellular apoptosis susceptibility protein, which is upregulated upon treatment of NCC with recombinant TNF. These findings suggest a parallel evolution of cell-mediated cytotoxicity in teleosts, leading to effector functions comparable to mammalian counterparts. INDEX WORDS: Cell-mediated Cytotoxicity, NK cells, Cytotoxic T Lymphocytes (CTL), Granule Exocytosis, Granzymes, Perforin, Granulysin, Tumor Necrosis Factor (TNF) alpha, Activation Induced Programmed Cell Death (AIPCD). PHYLOGENETICS OF EFFECTOR MOLECULES INVOLVED IN CELL-MEDIATED CYTOTOXICITY by KESAVANNAIR PRAVEEN M.S., University of Arkansas at Pine Bluff, 2001 M.F.Sc., Central Institute of Fisheries Education, India, 1998 B.F.Sc., Kerala Agricultural University, India, 1996 A Dissertation Submitted to the Graduate Faculty of The University of Georgia in Partial Fulfillment of the Requirements for the Degree DOCTOR OF PHILOSOPHY ATHENS, GEORGIA 2005 © 2005 Kesavannair Praveen All Rights Reserved PHYLOGENETICS OF EFFECTOR MOLECULES INVOLVED IN CELL-MEDIATED CYTOTOXICITY by KESAVANNAIR PRAVEEN Electronic Version Approved: Maureen Grasso Dean of the Graduate School The University of Georgia May 2005 Major Professor: Liliana Jaso-Friedmann Committee: Donald L. Evans David S. Peterson David J. Hurley Zhen F. Fu DEDICATION For my beloved wife Manju and my family. I am thankful for your unconditional love and support, without which this would not have been possible. iv ACKNOWLEDGEMENTS Heartfelt thanks to Dr. Liliana Jaso-Friedmann, my major professor and mentor, for being my constant source of guidance and encouragement all through this time. You have been above and beyond what I would have ever wished for as a mentor. I owe you a lot for all my achievements both at the professional as well as personal level. I am extremely thankful to my committee members, Dr. Donald Evans, Dr. David Peterson, Dr. David Hurley, and Dr. Zhen Fu for their continuous support, encouragement and invaluable suggestions. Many thanks to Dr. Dorothy Hudig, Dr. Dieter Jenne, Dr. James Powers, and Dr. Jan Potempa for their interest in my work, and for being there to offer helpful suggestions. Special thanks to John Leary for helping me with the experiments. Many thanks to Dr. Okinaga for sharing the reagents and protocols. Financial assistance from UGA's graduate school in the form of Dissertation Completion Assistantship is highly appreciated. I am quite fortunate to have wonderful friends like Jayakumar, Ritesh, Tina, Niraj, Naomi, Praveen, Neelesh, and Chen. My heartfelt appreciation for all their help and friendship. Words are not enough to express my gratitude and sincere appreciation for the Friedmann family for everything they have done for me. I feel really fortunate to have known you. I am obliged to Dr. Joseph Burleigh for his guidance and kindness. Special word of appreciation goes to Goodwin family for all their help and friendship. I would like to thank each and everyone in my family for their unconditional love and support. Finally I would like to express my sincere appreciation for the guidance and blessings from all my teachers. Your love and affection is my greatest strength. v TABLE OF CONTENTS Page ACKNOWLEDGEMENTS.............................................................................................................v LIST OF TABLES......................................................................................................................... ix LIST OF FIGURES ........................................................................................................................ x CHAPTER 1 INTRODUCTION .........................................................................................................1 2 LITERATURE REVIEW ..............................................................................................7 CELL-MEDIATED CYTOTOXICITY ....................................................................8 GRANULE-MEDIATED PATHWAYS OF CYTOTOXICITY............................11 CYTOTOXIC PATHWAYS MEDIATED BY TNF-SUPERFAMILY.................31 REGULATION OF CELL DEATH IN CYTOTOXIC CELLS .............................38 REFERENCES........................................................................................................41 3 EVIDENCE FOR THE EXISTENCE OF GRANZYME-LIKE SERINE PROTEASES IN TELEOST CYTOTOXIC CELLS ..................................................71 ABSTRACT ............................................................................................................72 INTRODUCTION...................................................................................................73 MATERIALS AND METHODS ............................................................................76 RESULTS................................................................................................................79 DISCUSSION .........................................................................................................84 ACKNOWLEDGEMENTS ....................................................................................87 vi REFERENCES........................................................................................................87 4 NONSPECIFIC CYTOTOXIC CELLS OF TELEOSTS ARE ARMED WITH MULTIPLE GRANZYMES AND OTHER COMPONENTS OF THE GRANULE EXOCYTOSIS PATHWAY......................................................................................106 ABSTRACT ..........................................................................................................107 INTRODUCTION.................................................................................................108 MATERIALS AND METHODS ..........................................................................110 RESULTS..............................................................................................................114 DISCUSSION .......................................................................................................119 ACKNOWLEDGEMENTS ..................................................................................124 REFERENCES......................................................................................................124 5 MOLECULAR CHARACTERIZATION AND EXPRESSION OF A GRANZYME WITH CHYMASE ACTIVITY FROM CYTOTOXIC CELLS OF AN ECTOTHERMIC VERTEBRATE ............................................................................141 ABSTRACT ..........................................................................................................142 INTRODUCTION.................................................................................................143 MATERIALS AND METHODS ..........................................................................145 RESULTS..............................................................................................................150 DISCUSSION .......................................................................................................156 REFERENCES......................................................................................................161 6 CONSTITUTIVE EXPRESSION OF TUMOR NECROSIS FACTOR-ALPHA IN CYTOTOXIC CELLS OF TELEOSTS AND ITS ROLE IN REGULATION OF CELL-MEDIATED CYTOTOXICITY.....................................................................190 vii ABSTRACT ..........................................................................................................191 INTRODUCTION.................................................................................................192 MATERIALS AND METHODS ..........................................................................194 RESULTS..............................................................................................................200 DISCUSSION .......................................................................................................204 ACKNOWLEDGEMENTS ..................................................................................209 REFERENCES......................................................................................................210 7 MOLECULAR CLONING OF CELLULAR APOPTOSIS SUSCEPTIBILITY (CAS) GENE IN OREOCHROMIS NILOTICUS AND ITS ROLE IN REGULATION OF NONSPECIFIC CYTOTOXIC CELL (NCC) FUNCTIONS .............................234 ACKNOWLEDGEMENTS ..................................................................................238 REFERENCES......................................................................................................238 8 SUMMARY AND CONCLUSIONS ........................................................................255 viii LIST OF TABLES Page Table 3.1: Oligonucleotide primers used in the cloning of catfish granzyme and for expression studies by RT-PCR......................................................................................................104 Table 3.2: Amino acid identity and similarity of mature catfish granzyme to other known granzymes and related proteins ...................................................................................105 Table 5.1: Organization of TLGR-1 gene....................................................................................188 Table 5.2: Pairwise comparison of TLGR-1 with related serine proteases .................................189 ix LIST OF FIGURES Page Figure 3.1: Cloning strategy used to identify and characterize the cDNA for catfish granzyme ..91 Figure 3.2: Compiled full-length catfish granzyme cDNA sequence............................................93 Figure 3.3: Multiple sequence alignment of predicted catfish granzyme sequence with known granzymes and related proteins .....................................................................................95 Figure 3.4: Phylogenetic analysis of catfish granzyme .................................................................98 Figure 3.5: Analysis of tissue expression of catfish granzyme....................................................100 Figure 3.6: Alignment of other fish granzyme sequences with catfish granzyme.......................102 Figure 4.1: Production of active and pro-CFGR-1 in Pichia pastoris.........................................127 Figure 4.2: Purification of recombinant proteins.........................................................................129 Figure 4.3: Enzymatic activity of recombinant CFGR-1.............................................................131 Figure 4.4: Correlation between cytotoxicity and exocytosis of granzymes with tryptase activity in catfish NCC .............................................................................................................133 Figure 4.5: Expression of other proteases in the cytotoxic granules of catfish NCC ..................135 Figure 4.6: Identification of multiple granzymes from catfish NCC...........................................137 Figure 4.7: Expression of other granule components in catfish NCC .........................................139 Figure 5.1: Genomic organization of TLGR-1 gene and promoter region ..................................167 Figure 5.2: Predicted three dimensional structure of TLGR-1 ...................................................170 Figure 5.3: Comparison of TLGR-1 with other granzyme sequences ........................................172 Figure 5.4: Phylogenetic analysis of TLGR-1 .............................................................................174 x Figure 5.5: Analysis of tissue expression of TLGR-1 .................................................................176 Figure 5.6: Transcriptional regulation of TLGR-1 in NCC.........................................................178 Figure 5.7: Production of recombinant TLGR-1 in Pichia pastoris............................................180 Figure 5.8: Purification of pro and mature TLGR-1 recombinant proteins .................................182 Figure 5.9: Enzymatic activity of recombinant TLGR-1.............................................................184 Figure 5.10: Substrate specificity of TLGR-1 .............................................................................186 Figure 6.1: Nucleotide and deduced amino acid sequence of tilapia TNF ..................................215 Figure 6.2: Multiple sequence alignment and molecular modeling for tilapia TNF ...................217 Figure 6.3: Phylogenetic analysis of tilapia TNF ........................................................................220 Figure 6.4: Expression of tilapia TNF gene.................................................................................222 Figure 6.5: Purification of recombinant tilapia TNF using Ni-affinity chromatography ............224 Figure 6.6: Expression of TNF on tilapia NCC membranes........................................................226 Figure 6.7: Cytotoxicity induced by tilapia TNF on susceptible WEHI cells .............................228 Figure 6.8: Recombinant TNF protects activation-induced cell death in NCC...........................230 Figure 6.9: Recombinant TNF upregulates the transcription of granzyme in tilapia NCC .........232 Figure 7.1: Compiled full-length cDNA sequence of tilapia CAS ..............................................242 Figure 7.2: Multiple sequence alignment of tilapia CAS ............................................................246 Figure 7.3: Phylogenetic analysis of tilapia CAS ........................................................................249 Figure 7.4: Expression of tilapia CAS in different tissues...........................................................251 Figure 7.5: Transcriptional regulation of tilapia CAS in NCC....................................................253 xi CHAPTER 1 INTRODUCTION 1 The significance of cell-mediated immunity became apparent with the discovery that professional killer cells were responsible for the clearance of virus infected and tumor cells and that these killing processes were not mediated though antibodies or complement. During the past few years, vast resources in time and effort were devoted to understanding the effector mechanisms used by those cytotoxic cells. The major players in cell-mediated cytotoxicity include cells of the adaptive immune system (cytotoxic T lymphocytes, CTLs) and natural killer cells (NK cells) that belong to the innate immune system. The molecular mechanisms by which these cells kill other cells have been investigated extensively and the importance of this process in innate and adaptive immune responses has been well established. Three stages have been identified in the cytolytic process mediated by professional killer cells. In the recognition stage, the target cells have to be identified by the effector cells. The second stage involves the lethal hit by which the effector cell kills the target cell. Lastly, the target cell disintegrates. Although some of the molecular mechanisms associated with these three crucial events have been identified, many more questions remain to be answered. Recognition of target cells by CTL involves presentation of foreign peptides in the context of class I MHC molecules and co-stimulators. Multiple receptors have been identified in target cell recognition by NK cells, and more are yet to be discovered. Following recognition, the signals initiated by the engagement of the receptors are a complex series of events that have not been fully deciphered, but are known to lead to the lethal hit of the targets. At least two independent pathways of killing have been identified in both CTL and NK cells: the granule exocytosis pathway with release of lethal molecules, and the binding of death ligands of the TNF superfamily to death receptors on target cells. It appears that the effector mechanism used by the cytotoxic cell varies with the type of target it has to destroy. Both pathways play significant roles 2 in inducing target cell death and can act synergistically, but the granule exocytosis pathway alone is sufficient to kill target cells even in the absence of death receptors. One of the aspects in the lethal hit that is not fully understood is the mechanism by which the cytotoxic activity is regulated in the activated killer cells. Much effort has been dedicated to understand the molecules involved in the killing of targets because, although CTL and NK cells employ different recognition mechanisms to identify their targets, the pathways to induce death are shared. Still, many questions remain to be answered about each of these pathways. Significant advances have been made in understanding the signaling pathways that are initiated by the ligation of TNF superfamily ligands to their corresponding death receptors. The granule exocytosis pathway is still poorly understood, due in part to the lack of information about the molecular identity of many of the granular components, as well as their physiological significance. Serine proteases of the granzyme family and other associated components of cytotoxic granules are the lethal components of the granule exocytosis pathway. Gene sequences for these molecules have been identified in mammals, but there is no such information available for coldblooded vertebrates. This fact represents a significant immunologic and evolutionary void in our knowledge, since granule exocytosis remains the preferred mechanism employed by T-cells and NK cells in the killing of tumor cells, protozoan parasites or virus-infected cells. One of the major obstacles immunologists and evolutionary biologists face in understanding the granule exocytosis pathway of killing is in trying to explain the role of multiple granzymes with overlapping substrate specificities. It is known that new highly specialized primary specificities are gained by recently evolved proteases, while the predicted evolutionary precursor already had very restricted substrate specificity. Thus, the evolution of 3 serine protease family members is perplexing and the events that have led to the despecialization steps in generation of newer specificities are still not clear. Granzymes are considered as newly evolved members of the serine protease family. They can be divided into four sub-families based on their specificity: Tryptase, Asp-ase, Chymase and Met-ase. Members of these subfamilies have co-evolved in separate genetic loci and their relative significance is not known. It has been well established that granzymes A and B induce target cell death, while the functional importance of other granzymes is unclear. These studies have been complicated by the realization that silencing of one or two granzyme genes alone does not inhibit all the cytotoxic activity of the effector cell. Clearly, simpler models of granule exocytosis killing mechanisms would aid in answering some of the fundamental questions about this pathway of cytotoxicity. Fish are the first group of animals in the evolutionary tree to develop a fully functional immune system with innate and adaptive immunity comparable to mammals. As such, fish represent an ideal model for the study of immunological processes in addition to understand unsolved questions regarding the development and function of the immune system in higher vertebrates. Moreover, the comparative study of different, evolutionarily distant fish species provides an excellent opportunity to understand the phylogeny of effector molecules. The first killer cell population of teleosts was identified and characterized in channel catfish and was called nonspecific cytotoxic cells (NCC). Subsequently, the presence of teleost cytotoxic cells other than NCC (as well as similar to NCC) has been reported from many species of fish. Initial observations suggested that NCC were non-granular. However, their dependence on calcium in inducing target cell death suggested the existence of the granule exocytosis pathway. Although the effector molecules involved in cytotoxic function and their regulation were not fully understood, killing of mammalian tumor target cells by NCC suggested that the 4 molecules involved in the lethal hit would be evolutionarily conserved. This theory was supported by functional studies that demonstrated the presence of components of the death ligand pathway in NCC from different species. While the role of Fas ligand in NCC cytotoxicity has been demonstrated, the functions of other members of the TNF super family remain poorly understood. Significance: The present study was undertaken to delineate the pathways of cytotoxicity employed by NCC in the delivery of the lethal hit. Identification of the components that mediate NCC function is important for several reasons. The identification of genes coding for components of cytotoxic granules from ectotherms would provide the first step in understanding the phylogenetic origin and the mechanisms of action of the many members of the granzyme family. This information could be used to understand the biological roles, substrates and pathways of death employed by different granzymes in organisms, where acquired immune responses appear to be less advanced. Aside from the basic knowledge in immunology gained by these studies, a practical application of this work is that characterization of the molecules involved in cell mediated cytotoxicity of teleosts will provide more insights into developing strategies to improve fish health. This is exemplified by the finding that unlike mammalian NK cells that can recycle and attack multiple targets, NCC lack the ability to recycle. It has been demonstrated that NCC undergo activation-induced cell death (AICD) following conjugate formation with the target cell. The role of death ligands as apoptosis regulatory factors has been suggested as a mechanism to balance the signals for AICD and cell survival. This mechanism could provide a way to enhance the NCC functions during episodes of acute stress. Hypothesis: The central hypothesis for this study was that NCC use pathways involving both granule exocytosis and TNF superfamily ligands to induce target cell death. This would 5 provide evidence for a parallel evolution of effector molecules in ectothermic vertebrates. The objective of this study was to identify and characterize the molecular components involved in NCC functions. To accomplish this, the following specific aims were pursued. 1 Identification of the components of cytotoxic granules in NCC from channel catfish (Ictalurus punctatus) and tilapia (Oreochromis niloticus) to compare the pattern of evolution in two phylogenetically distant species. This aim included: • Identification of granzyme gene sequences • Assessment of the expression pattern and substrate specificities of teleost granzymes • Identification of genes coding for accessory molecules present in cytotoxic granules 2 Identification of the role of death ligand TNF-alpha in functions of NCC in tilapia (Oreochromis niloticus). This aim included: • Identification of the tilapia TNF gene sequence • Assessment of the expression pattern of TNF in tilapia NCC (membrane and secreted forms) • 3 Functional characterization of TNF in tilapia NCC Identify the role of regulators of apoptosis in modulating NCC functions during episodes of acute stress. This aim included: • Identification of apoptosis regulatory genes expressed under acute stress • Assessment of their expression patterns 6 CHAPTER 2 LITERATURE REVIEW 7 CELL-MEDIATED CYTOTOXICITY The first report of the in vitro toxic effects of immune cells on target cells dates back to the 1960’s (1). In these early studies, the aims were to understand the physiological requirements for target cell death (2-4). Although lymphocytes were the prime suspects, the identity of the effector cells involved in these cytotoxic reactions was not fully understood. Subsequently, T lymphocytes were shown to play a major role in cell-mediated cytotoxicity (5,6). Cytotoxic lymphocytes are the professional killer cells that form conjugates with target cells prior to killing them. The target cells that are recognized by killer cells have been infected with cellular pathogens or are phenotypically transformed, as in the case of tumor cells. This group of cytotoxic effector cells includes cytotoxic T lymphocytes (CTL), natural killer (NK) cells, gamma delta T cells and natural killer T (NKT) cells (7). CTL cytotoxicity is triggered by TCR recognition of target cells bearing specific peptides on particular allotypes of MHC class I molecules. By contrast, NK cell cytotoxicity is regulated by a delicate balance between the engagement of inhibitory and activating germ line-encoded receptors. In spite of many differences, both NK cells and CTL use the same basic mechanisms for destroying their targets. (8). In NK cells, many of the effector molecules are pre-formed during development and they can initiate their cytotoxic function without undergoing proliferation. Unlike NK cells, CTL precursors are not cytotoxic and upon activation, they induce proliferation and upregulate the expression of effector molecules to become better killers (9,10). Cell-mediated cytotoxicity is important in regulation and termination of an immune response. The regulatory role is directed at inducing the death by apoptosis of activated immune cells. Uncontrolled cytotoxicity can lead to undesirable conditions like in the case of graft versus host disease (8). Target cell lysis by cytotoxic cells differs from the cytotoxicity induced by 8 antibody and complement in the fact that the former affects the nucleus resulting in internal disintegration of the targets (11,12). Target cells play an active role in their own destruction, which is obvious from the fact that the severity of nuclear destruction induced by the same killer cell varies with the nature of the target cell (13). The differential contribution of various effector molecules towards the killing is determined to a large extent by the quality of target cell used to measure the cytotoxicity (14). Nonspecific cytotoxic cells (NCC) are the first identified cytotoxic cell population in teleosts. Since their initial description in the channel catfish (15-18), these cells have been well characterized in a number of lower vertebrates (19-23). Cytotoxic cell populations with different phenotype than that of NCC have also recently been identified from channel catfish (24,25). The importance of the Fas ligand-mediated killing mechanism of mammalian target cells by fish cytotoxic cells has been previously reported (26-28). Although first described as agranular (15), NCC do have small granules and contact with target cells leads to granule exocytosis with necrotic and apoptotic death pathways in trout, tilapia and catfish (17,21,29). Requirement for calcium for the induction of cytotoxicity by NCC was an early indication that these cells use the granule exocytosis killing pathway (30). Catfish cytotoxic T cell lines have also been shown to kill target cells by the granzyme/perforin pathway, but the lack of molecular tools did not allow the identification of these proteins (31). NCC kill a variety of transformed mammalian cell lines, much like human lymphokine activated killer cells (LAK), which also kill xenogeneic targets. Unlike LAK cells however, NCC do not require activation with lymphokines to kill a wide range of targets. Target cell lysis occurs following conjugate formation of the target cell with an antigen receptor on NCC (NCCRP-1) (18,19). Complete abrogation or decreased expression of NCCRP-1 as well as its blocking with specific antibody results in inhibition of killing of target 9 cells (18,20). The phenotypic changes that follow cross-linking of the antigen receptor, NCCRP1 with its natural ligand on target cells or a monoclonal antibody (mAb 5C6) are signals for activation of cytotoxicity and degranulation (32). Effector mechanisms of cell-mediated cytotoxicity NK cells and CTLs can kill their targets by the vectorial release of the contents of their granules or by engaging death receptors on the surface of target cells with corresponding ligands (for example, the ligands of TNF superfamily). This generalization does not reflect the contribution of the multiple pathways involved, depending upon the type of effector molecule or the target cell, all of which will eventually lead to the cell death (10). The main constituents of cytotoxic granules are perforin and a family of serine proteases called granzymes (8). FasL and other similar TNF superfamily members are type II transmembrane proteins expressed mainly on T cells and NK cells (33). In non-hematopoietic cells FasL is directly sorted to the cell membrane (34). In contrast, FasL is co-expressed with granzymes and perforin in the secretory granules of NK cells and CTLs (35). The cytoplasmic tail of FasL contains sorting information, which targets newly formed protein to the secretory granules in cytotoxic cells (35). Fully active granule associated FasL is released along with granzyme and perforin to the immunological synapse between effector and target cells. This initial degranulation process, changes the expression of FasL to the cell membrane following target cell recognition (35). Although the different effector mechanisms used by cytotoxic cells are still not fully understood, most studies agree that there are two main killing pathways: the granule mediated pathways and the pathways involving members of the TNF superfamily. 10 GRANULE-MEDIATED PATHWAYS OF CYTOTOXICITY Cytotoxic Granules Upon formation of a well-defined immunological synapse between the target and effector cells, the contents of the cytotoxic granules are released in close proximity of the target cell (36,37). Professional secretory cells utilize secretory granules for exocytosis of their products while maintaining a separate lysosomal compartment. In contrast, hematopoietic cells have merged those two organelles in to a single compartment called the secretory lysosome (38). Secretory lysosomes resemble lysosomes in their low pH, similar structure and the marker proteins they contain (38-40). However, unlike lysosomes, they can be distinguished by their ability to fuse with the plasma membrane in a regulated fashion (41). The granules in cytotoxic cells have specialized structures with a core surrounded by a multivesicular region (39). Cytotoxic granules may play a dual role with a regulated secretory compartment (the core) and a pre-lysosomal peripheral compartment (42). Granzymes, a group of serine proteases comprise about 90% of the mass of these granules (43). Other components of the granule are perforin (39), calreticulin, which chaperons perforin (44), chemokines (45,46), chondroitin sulfate-containing proteoglycans called serglycins (47), granulysin (48) and some members of the TNF superfamily (35). The peripheral compartment of cytotoxic granules has several lysosomal hydrolases as well as unique components such as the mannose-6-phsophate receptor complex, usually absent in lysosomes, but present in early endocytic compartments (42). Among the lysosomal enzymes, cathepsin C (also called dipeptidyl peptidase I, DPPI) has functional significance in the activation of serine proteases. Other proteins identified in lytic granules include lysosomal transmembrane proteins (CD63), lysosome-associated membrane 11 proteins (Lamp1 and Lamp2) (39) and cytotoxic T lymphocyte antigen 4 (CTLA-4), which plays an important role in T cell signaling (49). Granzymes A total of 14 granzymes have been reported from humans and rodents that have been named A to M. Of these, granzymes A and B are the most abundant and the best studied to date (39). Reports of granzyme-like molecules from lower vertebrates are now becoming available (50). Structurally, granzymes are closely related to chymotrypsin, with conserved catalytic triad residues: His, Asp, and Ser (43). Granzyme subfamilies have trypsin-like, chymotrypsin-like or elastase-like specificities and their genes are mapped to tryptase, chymase or Met-ase loci, respectively (51). Most of the earlier studies on granzymes concentrated on granzymes A and B, while the importance of other granzymes (called orphan granzymes because of the lack of information about their physiological substrates) is now being widely appreciated (52). Like other serine proteases, the catalytic activity of granzymes depends on the Ser residue at the active site (51). There is an oxyanion hole to stabilize the transition state of the enzymesubstrate complex and a substrate binding pocket, which determines the substrate specificity. Few granzyme crystal structures have been resolved (50,53,54). The 3D structure data from other serine proteases has been used to construct molecular models that can predict the substrate specificity and structural features for many other granzymes. These models have helped in understanding the functional association of granzymes with other molecules and their predicted role in cell mediated cytotoxicity (55). Granzymes are synthesized as zymogens and are processed at the time of packaging into the granules. Cleavage of a leader peptide leaves two amino acids at the N-terminal and final 12 enzymatic activation is achieved when those two residues are clipped off by Cathepsin C (56). Granzymes A and B are found to have mannose-6-phosphate moieties, which will allow the packaging thorough the mannose-6-phosphate receptor pathway (57). The level of glycosylation varies widely between the granzymes. Granzyme K has no glycosylation, granzyme C has few and granzyme D is very heavily glycosylated (43,58). All granzymes do have three highly conserved disulfide linkages, which are responsible for the correct folding of the protein. Granzyme A, K and M have an additional disulfide bond bridging the active site serine, like in chymotrypsin (43). Granzyme A is the only granzyme found to form dimers as a result of interchain disulfide bonding (59). Cell specificity of granzyme gene expression has been the subject of many investigations (52). Granzymes were believed to be expressed mainly in CTL and NK cells (60). Initial analysis of granzyme expression in CTLs derived from mixed lymphocyte reactions revealed a predominance of granzyme A and B, with minimal expression of orphan granzymes (61). However, expression of orphan granzymes along with granzymes A and B was found to be high in NK cells. High level expression of orphan granzymes is restricted to lymphokine activated killer (LAK) cells, NK cells, large granular lymphocytes (LGL) and thymocytes. Both CD4+ and CD8+ T cells express orphan granzymes in an activation dependant manner, but at a very low level compared to granzymes A and B (52). Considering the high level of expression in thymocytes, it is hypothesized that orphan granzymes may play an important role in T cell development (62). Recent findings, using single-cell RT-PCR studies, have suggested that each lymphocyte expresses different combinations of perforin and/or one or more granzymes, indicating an additional level of control over lymphocyte mediated killing (63). 13 Recent reports suggest that granzymes could also be involved in non-immune functions (64,65). For example, granzyme genes have been found to be upregulated in cells treated with follicle stimulating hormone (64). The presence of perforin-independent expression of granzyme B and its specific inhibitor in human testis and placenta (66) could indicate a role of granzymes in reproduction. The release of granzymes during cytotoxicity causes an increase in the level of granzyme in the biological fluids and serves as a marker for activation of CTL and NK cells. Thus, soluble granzyme levels are found to be elevated in viral infections (67), parasitic infections (68), and rheumatoid arthritis (69). An alternatively spliced variant of granzyme K is expressed in certain neuronal cell types (70). Similarly, an alternatively spliced variant of granzyme M is exclusively expressed in photoreceptor cells of the retina in the mouse (71). The functional significance of these variants has not been fully investigated. Granule Exocytosis The granule exocytosis pathways of mammalian cells and the outcome of many of the complex events that take place in the killing of target cells have been recently reviewed (72). NK cells have high quantities of constitutive granules and thus are armed for immediate response following target cell contact. Once NK cells have killed a target, they require cytokine activation to recycle (73). In contrast, CTL require initial antigen stimulation for the up-regulation of cytokine receptor expression and for the synthesis of granular proteins (43). Subsequent encounters with specific antigen-bearing target cells provide the signals for the armed clones of CTL to degranulate and proliferate, a process that may take up to 3 days. This difference between NK cells and CTL may reflect the distinct roles of each one of these cells in the immune 14 system. NK cell activity is part of the innate immune system with rapid but limited cytotoxic activity. CTL take longer to become lethal, but acquire a higher killing capacity. Conjugate formation is initiated by the interaction of activation receptors with surface ligands on the target cells, although adhesion molecules are required to maintain a tight junction between the effector and the target cell. Redistribution of granules on the NK cell or CTL to the contact area with the target leads to vectorial release of granular contents and provides specificity of killing to the granzyme pathway. This is known as the "lethal hit" (74-76). The granules can move along the microtubule in both directions, towards and away from the microtubuleorganizing center (MTOC). Upon conjugate formation, the MTOC polarize towards the immunological synapse (77-79). The CTL and NK cell interactions with the target cells are very rapid (minutes) and transient, compared to the cell to cell interactions of CD 4+ T cells with antigen presenting cells (80). The degranulation required for cell-mediated cytotoxicity is controlled by actin polymerization, a requirement for killing (81-83). The delineation of the molecular mechanisms involved in the delivery of the lethal hit has been investigated extensively but are still not completely understood (84-87). As a result of these investigations, several important signaling molecules have been identified as critical for granule exocytosis such as Syk, Rac, Vav and ERK (81). These molecules appear to act as messengers that deliver signals from the receptor on the effector cell to downstream effectors that induce the cytoskeletal changes (88). It is important to note that the exact nature of the receptor(s) that initiate actin polymerization in cytotoxic cells to activate degranulation has not yet been confirmed. 15 Entry of granule components in to the target cells Models describing the entry of granule components into target cells have been undergone multiple revisions. Negatively charged sulfated GAG chains of proteoglycans act as a reservoir for cationic proteins found within the cytotoxic granules (40). During granule exocytosis, the contents of the granules experience a sudden change from highly acidic to physiological pH, which results in the release of perforin from the macromolecular complexes, while granzymes remain bound (40). Membrane pores formed by polymerization of perforin was initially believed to be the site of entry for granzymes into the target cells (89). The perforin pores fail to admit unglycosylated proteins or carbohydrate polymers and are found to be highly selective for granzymes (90). Other membrane pores, like the ones formed by complement, cannot effectively deliver granzymes into the target cells, suggesting a specific interplay between perforin and granzymes (90). However, fluorescein isothiocyanate (FITC) labeled granzyme B has been shown to enter the target cells even in the absence of perforin. It was demonstrated that granzyme B can bind to the target cell surface in a specific, saturable manner in the absence of perforin and remain in endocytic vesicles without any cytotoxic consequences (91-93). These findings suggested the existence of a specific receptor, which can initially internalize granzyme B into endocytic compartments. Sublytic concentrations of perforin, or any other endosmolytic agents like non-replicating adenovirus, listeriolysin O, or streptolysin O, can act to help the granzyme B to be released into the cytoplasm of target cells (90,91). While the mannose 6phosphate receptor has been reported as a candidate for cell surface receptor for granzyme B (94), recent reports highly contradict the significance of this receptor in induction of target cell death (95-97). 16 Once granzymes are released from cytotoxic granules, they are found as complexes bound in a charge dependent manner with the proteoglycan serglycin (98-100). Thus, new questions have been raised on the interaction of this complex with the receptor. Granzymes do have positive surface charge, which is responsible for the charge-dependant binding between granzyme and serglycin. The masking of charges due to this binding is predicted to affect the interaction between granule components and cell-surface receptors on target cells (53,101,102). Several candidate proteins have been reported to be involved in the uptake of the granzymeserglycin complex. Dynamin (103) as well as cell surface-bound heat shock protein-70 have been reported to bind to and uptake granzyme B, resulting in a perforin-independent target cell death (104). Role of perforin in cytotoxicity Cytotoxic lymphocytes have been described to form conjugates with their targets and deliver a “lethal hit”, resulting in target cell death (105). Perforin, a major component of the cytotoxic granule, is synthesized as an inactive precursor, which will undergo proteolytic processing to remove carboxy-terminal glycosylated peptide and become active (106). Active perforin monomers undergo calcium-dependant polymerization and get inserted into the target cell membrane, forming pores on cell surfaces like the C9 component of terminal complement complex (107). Target cell destruction was believed to be a result of osmotic lysis due to excessive influx of ions into the target cells (108,109). Although gene knock-out experiments have shown that perforin is required for target cell death (110-113), the exact mechanism remains unclear. The discrepancy is partly due to the fact that the hallmark of target cells attacked by cytotoxic lymphocytes is apoptosis, but perforin alone induces membrane damage 17 leading to necrosis (114,115). These observations led the way to the hypothesis that granzymes have an important role in target cell death (116). The exact function of perforin in delivering granzyme to the target cells is still under debate. In some circumstances perforin alone might bring about the target cell death (90,99). Some of the main problems associated with determining the role of perforin in cytotoxicity is the failure to generate active recombinant perforin and inability to track perforin movement inside target cells by fluorescent microscopy (8). In an effort to clarify the respective roles of perforin and granzymes in target cells death, these components have been added separately in transfection experiments. When the non-cytotoxic rat basophilic leukemia (RBL) cell line was transfected with perforin, high level of cytolysis against RBC was observed, while nucleated cells were not killed effectively (117). These results appear to indicate the poor efficiency of perforin in forming pores in nucleated cells. A possible explanation could be that nucleated cells have the capacity to repair membrane damage (118). Transfection of both granular components, granzyme and perforin, produced apoptosis in nucleated target cells (91). Role of granzymes in cytotoxicity The first evidence for the involvement of cellular proteases in cell-mediated cytotoxicity came from the experiments involving inhibition of killing by T-cells with general protease inhibitors like diisopropylfluorophosphate (DFP) and phenylmethylsulfonyl fluoride (PMSF) (119-122). Other natural protease inhibitors like alpha1-antitrypsin and alpha1-antichymotrypsin can also suppress target cell killing (123,124). Pre-treatment of effector cells with these protease inhibitors had no effect on the target cell killing, but the presence of inhibitors during the effector-target conjugate formation, even for a short period of time, was found to be crucial in 18 inhibition of cytotoxicity (125). Similarly, a short period of incubation with microfilament inhibitors was crucial in blocking target cell death (125). These results suggested that secretion of proteases to the extracellular environment in close proximity to the target cells was a necessary step in cell-mediated cytotoxicity. Subsequently, a DFP sensitive protease was isolated from cytotoxic T cells and was found to be lethal to T24 human bladder carcinoma cells, suggesting the direct involvement of such proteases in cytotoxicity (126). The serine proteases present in cytolytic granules of cytotoxic lymphocytes were named granzymes. Granzyme B: Apoptosis induced by CTLs is associated with the proteolytic processing of procaspase-3 (127). The most studied of all the granzyme family members, granzyme B, was originally thought to act by cleaving caspases and inducing apoptosis. Subsequent studies on the hierarchy of granzyme B-initiated caspase activation suggested a two-tiered activation cascade, involving seven caspases (128). More recently, granzyme B has also been found to induce a caspase-independent pathway of cell death (129,130). Indeed, multiple intracellular substrates for granzyme B have been identified, which are associated with cell death pathways. Two of the alternative mechanisms of action of granzyme B are the direct cleavage of caspases to induce DNA fragmentation (131-133), or the cleavage of key structural proteins in the nuclear membrane or cytoskeleton (134,135). One of the most important direct targets for granzyme B is BH3-interacting domain death agonist (BID), which following truncation can destroy the membrane integrity of mitochondria (136). Mitochondrial lysis leads to the release of multiple pro-apoptotic molecules including cytochrome c, HtrA2/OMI (a serine protease that blocks inhibitor of apoptosis proteins) and endonuclease G (137-140). However, there is also evidence for the disruption of mitochondrial transmembrane potential by granzyme B, which is independent of BID and caspase (137,141,142). Therefore, it can be hypothesized that there are 19 multiple caspase-independent mitochondrial effects of granzyme B, one of them requiring BID and the other one involving direct action of granzyme B (8). It has been demonstrated that the truncated BID generated by granzyme B proteolysis uses Bak, a mitochodrial proapoptotic Bcl-2 family member, for the release of cytochrome c from mitochondria. These results have been derived from the lack of granzyme B-mediated cytotoxicity in Bak deficient target cells (143). Furthermore, these data appear to concur with the fact that Bak deficiency has been hypothesized as an immune evasion mechanism used by certain tumor cells and virally transformed cells. Mitochondrial apoptotic events initiated by granzyme B can occur even in the absence of BID, indicating alternative pathways (144). The alternative mechanism involves Bim, a member of the BH3-only-protein family, which can mediate the release of cytochrome c. Mcl-1L, which is a pro-survival protein, can bind to Bim and mask its pro-apoptotic effects. Mcl-1L localizes mainly to the outer membrane of mitochondria and is found to be a physiological substrate for granzyme B (144). These findings suggested a novel pro-apoptotic pathway induced by granzyme B. Inhibition of Mcl-1L in cells can induce apoptosis even in the absence of any apoptotic stimuli, indicating the significance of this pathway in cell death (145) Additionally, other pro-apoptotic effects of granzyme B are believed to be due to the cleavage of downstream caspase substrates like poly (ADP ribose) polymerase, catalytic subunit of DNA-dependant protein kinase and nuclear mitotic apparatus protein (146,147). A recently identified pathway involves the cleavage of Rho-associated coiled coil-containing protein kinase (ROCK) II by granzyme B (148). ROCK I is a caspase substrate and its breakdown leads to the membrane blebbing in apoptotic cell death. ROCK II is closely related and has similar functions 20 to ROCK I. Cleavage of this protein by granzyme B indicates yet another caspase-independent pathway to induce apoptotic morphology in target cells (148). Other substrates for granzyme B include the zeta chain of T cell receptor (TCR), which is cleaved at a different aspartic acid residue than that of the cleavage mediated by caspase-3 (149). It has been shown that viruses like HIV can use the TCR zeta chain to contribute towards the pathogenesis as an evasion mechanism (150,151). Therefore, direct degradation of the TCR zeta chain by granzyme B has been hypothesized as a cytotoxic mechanism directed against T cells that have been infected with such viruses. Granzyme A: Most of the initial studies on granzymes concentrated on granzyme B because it was fast and easy to measure its action on target cells, inducing classical signs of apoptosis. Until recently, granzyme A was believed to be an inert granzyme or a very slow killer (8). This idea was due to the absence of apoptotic signs, such as oligonucleotide fragment release, for up to 16 hours post-treatment. Transfection experiments done with the RBL cell line have shown that granzyme A, in combination with perforin, induces apoptosis in a perforindependant manner (152). It is now apparent that recombinant granzyme A can induce cell death as rapidly as recombinant granzyme B, but in a caspase-independent way (153). Cell death induced by granzyme A has many of the classical features of apoptosis, including chromatin condensation, externalization of phosphatidyl serine on cell membrane, loss of mitochondrial transmembrane potential and nuclear fragmentation (134,153). Curiously, granzyme A is not active on any of the downstream caspases. In addition, the loss of mitochondrial membrane potential did not result in release of cytochrome c and overexpression of Bcl-2 did not have any observable effects on the granzyme A mediated killing (8). 21 A set of molecules targeted by granzyme A have been identified. The main granzyme A target is an endoplasmic reticulum-associated complex called SET (154). The principal components of this complex include two tumor suppressor proteins (pp32 and NM23-H1), the nucleosome assembly protein SET, the DNA-binding protein high-mobility group protein-2 (HMG-2), and the rate-limiting base excision repair enzyme apurinic/apyrimidinic endonulease 1 (APE-1) (155-157). Although the exact role of the SET complex is still unknown, it has been hypothesized that it acts to facilitate activation of transcription and translation-related DNA repair in response to oxidative stress (157). Granzyme A cleaves SET, HMG-2 and APE-1 in a perforin-dependant manner. The DNA damage in response to granzyme is different than that induced by granzyme B. Granzyme A induces single stranded nicks while oligonucleosomal DNA fragments of ~200 bp are generated by granzyme B. The SET complex is believed to be the key factor in the induction of DNA nicks. NM23-H1 is a granzyme A-induced DNAse (GADD), while SET is its inhibitor (IGADD) (158). NM23-H1 has been shown to nick DNA (159). After the granzyme A attack, SET and NM23-H1 translocate to the nucleus. While SET has been degraded by granzyme A it allows for the DNAse activity of NM23-H1 to nick DNA. Granzyme A and B target some important nuclear proteins. Both granzymes A and B are found to translocate to the nucleus soon after entering the cells (160,161). Lamins are important structural components of the nuclear envelop and they form the targets for both granzyme A and B (134). Disruption of the nuclear envelop is believed to be a key step in apoptosis. Granzyme A also targets linker histone H1 (162). Degradation of histone H1 facilitates the opening up of the chromatin, making it more accessible for granzyme B. Orphan granzymes: Granzyme K is believed to be very similar in activity to granzyme A and is thought to be responsible for the residual esterase activity (Z-Lys-SBzl) of cytotoxic cells 22 from granzyme A deficient mice (163). Generation of recombinant granzyme K and resolution of its structure have been reported (54,58,164). However, there are no documented studies for the cytotoxic effects of recombinant granzyme K. Purified native granzyme K induces caspaseindependent cell death, without classical apoptotic nuclear morphology. Disruption of mitochondrial membrane potential was observed in target cells in response to granzyme K (142). Recombinant granzyme H is reported to have chymase activity, but the cytotoxic effects of this granzyme has not been reported (165). Another member of orphan granzyme family is granzyme M, which is of particular interest due to its preferential expression in cytotoxic cells of innate immunity including NK cells and gamma delta T cells (166). Prediction for a role of granzyme M in target cell lysis with sub-optimal level of perforin has not been successful (51), but other roles have been assigned to granzyme M. One of the hypothetical roles for granzyme M that has been put forward suggests that it could be responsible for damaging viral particles during the lysis of infected cells (166). While that mechanism is not well understood, the role for granzyme M as a regulatory protease has been demonstrated (167). It has been shown that proteinase inhibitor 9 (PI-9) is a major inhibitor of granzyme B (168), and recent findings suggest that granzyme M is not only resistant to PI-9, but that it can effectively hydrolyze the inhibitor (167). Over expression of PI-9 has been reported as one of the evasion strategies used by certain tumors (169). Thus, combined delivery of granzyme B and M in target cells expressing PI-9 could facilitate the breakdown of the inhibitor by granzyme M, allowing the active granzyme B to induce target cell death through multiple pathways. In addition to the regulatory roles for granzyme M, a novel form of perforin-dependant cell death mediated by granzyme M has been recently reported (170). The kinetics of 23 cytotoxicity induced by granzyme M was comparable to that induced by similar amounts of granzyme B, indicating the importance of this novel pathway. In contrast to granzyme A and C, cell death induced by granzyme M did not involve the mitochondrial pathways. Overexpression of Bcl-2 had no effect on the killing. Unlike granzyme B, cell death mediated by granzyme M lacked the DNA fragmentation and occurred independent of caspase activities. These results suggest that granzyme M-mediated cell death is molecularly distinct from all known pathways (170). It is logical to extrapolate these findings to other orphan granzymes also, which might be using still unidentified cell death pathways. As explained above, cell-mediated cytotoxicity is a tightly regulated process. An additional major component of cytotoxic granules is calreticulin (171). Calreticulin is known as an inactivator of perforin-mediated lysis (171,172). One of the orphan granzymes, chymase 1 is reported to be the “inactivator of inactivator” because of its ability to cleave calreticulin and release active perforin (173,174). In vivo significance of granzyme-mediated killing The role of granzymes in inducing target cell death has been investigated extensively in vitro and it is better understood than the role of perforin in cytotoxicity at the molecular level (175). However, advances in in vivo studies demonstrating the significance of individual granzymes in cytotoxicity lag behind that of perforin. Mice deficient in one or more granzymes have focal immune deficits unlike the case with perforin knock-out mice, which have a wide spread inability to kill target cells (175). Since many of the genes encoding granzymes are structurally related and highly linked, deletion of one granzyme can result in the disruption of the expression of other granzymes (61). 24 Cytotoxic cells isolated from granzyme A deficient mice retain the potency to induce apoptosis of target cells in vitro (176). The residual cytotoxicity could be partially attributed to the intact serine esterase activity of granzyme K that remains in granzyme A-/- mice (163). Cytotoxic cells from granzyme B deficient mice can still exhibit classical signs of apoptosis in target cells, but at a slower rate than the wild type (177). As granzyme B cluster deficiency in mice is characterized by the additional loss of granzyme C and other neighboring orphan granzymes (61), these findings suggested that the in vivo effects of cytotoxic cells are not solely dependent on the granzyme B cluster. However, simultaneous disruption of granzymes A and B functions resulted in increase in susceptibility towards ectromelia virus infection (178). Recent attempts to understand the individual effects of members of the granzyme B gene cluster have revealed the significant role played by orphan granzymes present downstream of the granzyme B gene in mice (179). Both lymphokine activated killer cells and CTLs from mice deficient in the entire granzyme B cluster had more severe cytotoxic defects than those from mice deficient for granzyme B only (179). The emerging picture from recent studies points towards indispensable, simultaneous roles of multiple granzymes rather than in isolation in controlling certain viral infections (111,178,180). However, this pattern of susceptibility is not true in all viral infection models, suggesting that granzymes might have some additional extracellular functions or they might have evolved specifically to restrict replication of specific viruses (181). Synthetic Substrates and Inhibitors Granzymes are broadly classified by their primary enzyme specificity. This specificity is determined by the recognition of substrate residues immediately N-terminal to the sessile bond 25 (P1 residue). Based on the three dimensional structures of the members of serine proteases, it has been proposed that three residues of the S1 substrate binding pocket confer the primary specificity for all proteases. These residues are located at positions 189, 216 and 226 (chymotrypsin numbering) of the proteases (182). Four different enzymatic activities have been described for all the known granzymes and they are: Tryptase, Met-ase, Asp-ase and Chymase. Granzymes A and K have Asp at position 189, which permits ionic interaction with Arg or Lys at position P1. This arrangement indicates that granzymes A and K have tryptase activity (183). In granzyme B, the arginine residue at position 226 at the back of the primary binding pocket (S1) interacts with Asp or Glu at the P1 position, facilitating highly specific Aspase activity (184,185). The metase activity of granzyme M is determined by two residues, Lys-179 and Ser-201. Mutations of these key residues significantly reduced the Metase activity of the enzyme, but had chymase activity towards Phe at P1 site (186). Although granzyme C has Gln at position 226, which would predict preference for polar residues like Asn or Ser at P1 position, its actual enzymatic activity remains unknown. Furthermore, while granzymes D-H have similar residues around the S1 pocket predicting preference for Phe/Leu residues at P1 site (183), only granzyme H has been shown to have chymase activity. Synthetic peptide substrates are commonly used to detect enzyme activity during isolation and to determine the specificity. The three commonly used synthetic substrates for serine proteases are peptide thioesters, peptide p-nitroanilides (pNA) and peptide derivatives of 7-amino-4-methylcoumarin (AMC) (183). Compared to pNA and AMC, thioester substrates are more sensitive and thus more useful in characterizing new and unreactive proteases (187). Hydrolysis of thioester substrates can be monitored spectrophotometrically in the presence of 5,5’-dithiobis(2-nitrobenzoic acid) (DTNB) to determine enzyme kinetics (188). A library of 26 Boc-Ala-Ala-X-Sbzl (X = Various P1 residues) along with Z-Lys-Sbzl, Z-Arg-Sbzl and Suc-PheLeu-Phe-Sbzl have been used to study the activities of various granzymes (173,189-193). Synthetic inhibitors are powerful tools for identification of specificity and determination of in vivo and in vitro functions of proteases. There are five major classes of inhibitors used to study granzyme functions: simple substrate analogs, transition state analogs, alkylating agents which react with active site histidine, acylating agents which react with active site serine and mechanism-based (suicide) inhibitors (183). The first two classes of inhibitors are reversible while others are irreversible. While general serine protease inhibitors like PMSF and DFP have been used to identify granzymes as serine proteases (194,195), more specific inhibitors have been developed to study specificity and function of granzymes. Irreversible inhibitors, such as substituted isocoumarins, peptide chloromethyl ketones, peptide phosphonates, and guanidinobenzoate derivatives have been shown to differentially inhibit granzymes A, B, K and chymase I (173,192,195-197). Evolution of granzymes Functional redundancy of granzymes suggests the need for multiple molecular pathways to compete with viral and tumor strategies to evade cell death. These pathogens have coexisted and coevolved with immune systems of organism for millions of years (175). For example, adenoviruses resist killing by granzyme B by producing pseudosubstrates (198). Discovery of nonredundant mechanisms of action of granzyme A, B and C indicates importance of multiple granzymes in providing a “fail-safe” mechanism for anti-viral and anti-tumor immunity (8,199,200). This is exemplified by the ATP-independent cell death induced by granzyme A, that could be the result of a race between the host and the virus for energy sources (201). When the 27 energy resources for the cell are depleted, the caspase-dependant apoptosis is blocked (202) and viruses could have used this as an evasion strategy. The immune system appears to have developed alternative pathways, like the one used by granzyme A to trigger cell death under such conditions (203). Evolution of serine proteases is believed to be a result of mutations in the S1 pocket residues of the gene. However, attempts to interchange the primary specificity between the major superfamily members through appropriate chimeric replacement of S1 pocket residues were found to result in complete abolition of activity (204,205). It is believed that the ancestral serine protease family members were resistant to inactivation due to mutational changes to S1 pocket residues because of the structural plasticity of the S1 subsite, as evident in invertebrate and prokaryotic serine proteases (206,207). However, these proteases have broad multivalent primary specificities, suggesting progressive narrowing of primary specificities for proteases during vertebrate evolution. Once a narrow specificity is reached, further generation of diversity is achieved only by reversing to the original state (208). Mammalian granzymes are separated into clusters on three chromosomes and there is considerable sequence similarity among the members from the same group (52). There is ample evidence suggesting a gene duplication of granzyme B leading to the evolution of granzyme C in mice and granzyme H in human. Similarly, granzymes A, K and M seem to be related to each other. Early gene duplication of granzyme A could have led to the evolution of granzyme K (52). These two groups are believed to have evolved before the divergence of human and rodents. However, a third group consisting of all other orphan granzymes is believed to have evolved in rodents after such a divergence (52). 28 Role of granulysin in cytotoxicity Granulysin is a member of the saposin-like protein family that includes amoebapores and NK lysin (209). Expression of granulysin is found to be restricted to CD4+ and CD8+ T-cells, NK cells and gamma-delta T cells (210-212). Granulysin was found to be upregulated in cytotoxic T lymphocytes following activation and was shown to be cytotoxic against microbes and tumor cells (48,213,214). The three dimensional structure of granulysin has provided new insights into a possible mode of cytotoxicity for this molecule (215). The model predicts that the positive charges on granulysin could help the molecule to orient towards negatively charged surface molecules of target cells. Clusters of granulysin could cause lysis of the membrane by rolling in the direction of its first, second and third helices, creating a scissoring motion. This would further expose the lytic surface allowing granulysin to enter deeper into the target cell, bending or tearing it (216). CTL cytotoxicity against Cryptococcus neoformans was recently demonstrated to be dependent on granulysin expression, but not on the expression of perforin (48). In contrast, perforin was found to aid in the granulysin-mediated lysis of Mycobacterium tuberculosis, indicating the need for a membrane perturbing agent to facilitate the access of granulysin to an intracellular organism (48). The effective induction of apoptosis by granulysin of virally infected cells, like in the case of varicella-zoster infection, has been reported (217). This protection appears to be virus specific as granulysin had no effect on HIV replication (218). Another important immune effector mechanism of granulysin is as an effective cytotoxic agent against a variety of human tumors (219). Recent reports suggest that granulysin can be used as a marker for the outbreak of certain human cancers. Progression of cancer was found to be associated with decreased expression of granulysin in NK cells, while there was no correlation between tumor 29 progression and expression of perforin in these cells (220). In animal tumor models, efficacy of granulysin as a therapeutic agent has been demonstrated. By expressing granulysin using adenoviral vectors in a mouse lung cancer model, significant induction of apoptosis in tumor cells was achieved (221). Regulation of granule-mediated killing Cytotoxic lymphocytes have developed mechanisms to protect themselves from the lethal effects of granules they secrete (222). Surface expression of cathepsin B plays a significant role in protecting the killer cells from accidental cell death. Cathepsin B can cleave perforin, preventing the pore formation and protecting from membrane damage (223). Inactivation of cathepsin B using membrane impermeable inhibitors produced perforin-dependant suicide of CTLs and NK cells upon degranulation triggering (223). Cytotoxic lymphocytes also express an abundance of endogenous serpins, such as protease inhibitor-9 (PI-9), which would act as a pseudosubstrate for granzyme B and protect the killer cells (168,224). Serpins were first discovered as blood plasma proteins regulating fibrinolysis, blood coagulation, complement activation and tissue modeling (225). Plasma serpins like alpha-1 protease inhibitor and antithrombin III can inhibit granzyme A and B (226,227). Expression of PI-9 in immune privileged sites suggests its role in protection of bystander cells from destruction (228). Some types of tumor cells can escape the immune system by upregulating PI-9 expression (169). Another known granzyme inhibitor that protects target cells against granzyme B mediated killing is a viral inhibitor called cytokine response modifier (Crm A). Cowpox Crm A has been shown to inhibit both cysteine and serine proteases very 30 effectively (168,229,230). Other serpins such as SPI-6, mBM2A and raPIT5a, expressed in pituitary gland are also effective inhibitors of granzymes (231,232). CYTOTOXIC PATHWAYS MEDIATED BY TNF SUPERFAMILY Tumor necrosis factor Tumor necrosis factor and lymphotoxin were first identified as products of lymphocytes and macrophages that caused lysis of certain tumor cells (233,234). Later, more proteins with similar structure were identified and all of them are now classified as members of the TNF superfamily (235). The TNF-related ligands are type II transmembrane glycoproteins (Cterminus on the exterior, single transmembrane domain and short cytoplasmic N-terminus) with conserved C-terminal domain called TNF homology domain (236). Members of TNF superfamily have unique structural features that couple them directly to signaling pathways for cell proliferation, survival and differentiation, enabling them to assume prominent roles in coordinating proliferation and protective functions of pathogen-reactive immune cells (237). The TNF homology domains are -sandwich structures containing two stacked each formed by five anti-parallel pleated sheets strands that adopt a “jelly-roll” topology (238,239). The inner sheets are responsible for the trimer contacts while the outer sheets are exposed to the surface. The oligomer formation is crucial for TNF functions because the receptor binding sites on TNF are located at the interfaces of adjacent subunits. The TNF homotrimer can aggregate up to three receptors (240). TNF is regarded as a major player in orchestrating differentiation and proliferation as well as induction of cell death. The role of TNF in various pathological conditions, like cachexia, septic shock, rheumatoid arthritis and autoimmunity has been well established (241). 31 TNF and cytotoxic cells Antigen-specific CTLs have been shown to express members of the TNF ligand superfamily on their cell surface and/or as a secreted form (35,242-248). The role of these ligands in initiating cell death pathways in target cells were demonstrated with the use of specific blocking reagents, which disrupt the interaction between the ligand and corresponding receptor (245,247,249-251). Among various TNF superfamily members, Fas ligand is the one which is most intensively studied for its role in induction of cytotoxicity. All activated immune cells are found to express TNF alpha on their cell surface and secrete it as a soluble form (242,243,245,252,253). Expression of TNF in T cells is rapidly inducible by of the T-cellactivating stimuli, but the expression is transient, lasting a few hours (242). Freshly isolated (non-activated) and activated NK cells express a variety of TNF superfamily ligands, both at the level of mRNA and protein (254). NK cells have been shown to express TNF both on the cell surface and in the cytoplasm. Activation of these cells resulted in an increase in the expression of TNF-mRNAs, but not membrane protein. (254). However, activated NK cells showed significant increase in the secretion of soluble TNF (255). TNF-induced target cell death The two physiological forms of TNF (26 kDa membrane-bound form and 17 kDa secreted form) are effective in killing tumor cells and virally infected target cells upon cell-tocell contact. An uncleavable mutant version of TNF was found to be very effective in inducing cytotoxicity, suggesting that internalization of TNF is not a requirement for the cell death (256). The secreted form of TNF is generated by proteolytic cleavage of the membrane-bound form by a metalloprotease called TNF converting enzyme (TACE), which is expressed on the cell surface 32 (257). Both membrane-bound and secreted forms of TNF have been shown to be the major cytotoxic effector mechanisms used by macrophages and CTL (243,258,259). The action of membrane-bound TNF is considered to be more localized, requiring the cell-to-cell contact between the effector and target cells, compared to the more systemic effects of the secreted form of TNF (241). The contribution of TNF towards the killing mediated by lymphocytes was not fully appreciated before due to the dominant effects of perforin and Fas ligand in acute cytotoxicity (260). After the derivation of null mice for perforin (110), TNF (261) and the description of FasL mutant gld mice (262,263), the significance of TNF alpha in cell-mediated cytotoxicity was better understood. All of the cell-mediated cytotoxicity can be described as the end result of one or a combination of three mechanisms: pathways involving perforin/granzyme pathway, Fas/FasL or TNF/other similar ligands (264). LAK cells generated from perforin and FasL deficient mice were not cytotoxic in short term cytotoxicity assays, indicating the role of those two molecules in acute cytotoxicity. However, upon prolonged incubation with TNF-sensitive targets, these cells were effective in inducing cell death, which was blocked by addition of antiTNF antibodies. Both membrane-bound and soluble forms of TNF were effective in killing (253). T-cell receptor activated cloned CD8+ CTLs have been shown to express both the membrane-bound and the secreted forms of TNF in high quantities. TNF was found to be biologically active and the expression levels were highly upregulated upon activation of the cell, indicating in vivo significance of TNF-alpha in a slower CTL-mediated cell death pathway (259). Similarly, an ovalbumin peptide-specific CTL clone was found to use two distinct mechanisms of cytotoxicity. One of them was obvious within four hours while the other took two days and 33 was neutralized by anti-TNF antibodies (265). Perforin-independent killing of certain targets by CTLs was shown to be entirely due to TNF (266). These studies have demonstrated that the lymphocytes choose from the three pathways to suit a particular situation according to the susceptibility of the target and even effector to target ratios (267). Antigen-specific target lysis by CTL also results in some degree of bystander cell death, which is independent of class I molecules. Most of the bystander target lysis is attributed to the Fas-FasL interactions between the activated effector and non-antigen presenting targets (268,269). There was a significant reduction in the extent of bystander cell death by CTLs from gld mice (270). However, after prolonged incubation (for 36-48 hours), delayed bystander lysis was observed, which was shown to be mediated though the soluble form of TNF-alpha. There is no evidence for the induction of bystander cell death by soluble FasL. These results indicate that CTLs might use FasL or TNF in a kinetically and physically distinct fashion to mediate bystander cell death (270). Role of TNF as a bystander in the clearance of certain kinds of tumors also has been reported (271,272). The importance of Fas-FasL interaction in mature T-cell death has long been established (273). Many studies have suggested a Fas-independent mechanism, which also might play an important role in activation-induced cell death (AICD) pathways (274,275). Studies using lpr mice demonstrated that activation-induced cell death in response to a strong stimulus was comparable to that of normal T cells (276). Treatment of T cells with anti-TNF antibody or injection of antisense TNF oligodeoxynucleotides was shown to have an inhibitory effect on AICD in those cells (277). Later, TNFR2 was found to be responsible for Fas-independent activation-induced cell death of CD8+ CTLs (278). In addition to TNFR2, TNFR1 also is believed to be involved in AICD (at least in a CD8+ population of transgenic mice) where Fas- 34 independent pathways were found to be important (279). AICD in CD4+ T cells was believed to be entirely due to Fas-FasL interaction (280). However, TNF also was shown to play an additional role in this pathway (281). A combination of lpr phenotype and deficiency of TNFR1 had an accelerated lymphoadenopathy and increased serum immunoglobulin levels (282). These studies suggest that TNF-TNFR interactions can compensate for the loss of Fas-FasL signaling pathway in inducing AICD in T cells. Non-lytic functions of TNF In addition to a role in cytotoxicity, TNF-TNFR interactions can result in cell activation and proliferation. Induction of these biological responses appears to depend on TRAF-mediated triggering of kinase-dependant signaling pathways, leading to activation of transcription factors like NF-κB and AP-1 (264). Activation of these transcription factors can induce the expression of genes like inhibitor of apoptosis proteins (IAPs), which can inhibit cell death induced by TNF (283). It has been demonstrated that cIAP-1 and cIAP-2 can inhibit TNF-mediated activation of downstream caspases (284,285). Cells that produce TNF are generally responsive to TNF, suggesting autocrine feedback loops. However, the role of the membrane bound form of TNF in such feedback loops is not fully understood. Cells that permanently express membrane-integrated mutant version of TNF are found to express TNF-TNFR complexes on their surfaces (286). These cells were shown to have permanent activation of NF-κB, constitutive p38 mitogen-activated protein kinase activity and increased expression of IL-6. These results suggest that cells that express both TNF and TNFR on their surface can initiate autocrine signaling loops that might be important for their optimal activation and survival. The apoptosis-inducting capacity of soluble forms of TNF- 35 superfamily ligands has been shown to be reduced significantly, compared to the membranebound form (287-290). Recombinant soluble TNF-α and lymphotoxin are capable of inducing apoptotic cell death in mouse as well as human T lymphocyte blasts in vitro (291). Another study showed that membrane-bound FasL was cytotoxic for human peripheral blood lymphocytes, while the soluble form blocked the killing (292). TNF superfamily ligands can enhance T cell growth though coactivation signaling along with TCR signaling or other activation stimuli (293). Proliferation of gamma-delta T cells was also reported in response to TNF (294). The proliferative response triggered by a mixture of antiCD2 monoclonal antibodies or LFA-3 was found to be enhanced by the addition of TNF (295,296). Combination of TNF and IL-6 has been shown to have highly effective synergestic signals for T cell proliferation (297). TNF-induced signaling pathways leading to cytotoxicity or survival Ligation of TNF ligands induces oligomerization of the receptors on the target cell surface resulting in the aggregation of cytoplasmic domains. Formation of this complex is the first step in a series of signaling events inside the target cell, leading towards cell death (298). Based on binding of a variety of adapter molecules to the cytoplasmic tail of TNFR, a conserved domain called death domain (DD) was identified, which is necessary to initiate apoptosis in target cells. Another conserved domain identified is the one which binds to TNFR-associated factors (TRAFs). Binding of TRAFs to the multimerized TNFRs activates signaling pathways leading to cell death. A series of events have been characterized, which eventually lead to the cleavage and activation of caspases and to apoptosis. Alternatively, activation of TNFR can 36 result in kinase-dependant signal transduction and the activation of transcription factors that are crucial in regulation of cell activation and survival genes (298,299). The first protein to be recruited to the multimerized TNFR1 is TNFR1-associated death domain protein (TRADD), which serves as a platform for the recruitment of multiple proteins like receptor-interacting protein (RIP), RIP-associated ICH-1 homologous protein with a death domain (RAIDD), Fas-associated death domain protein (FADD) and TNF receptor-associated factor-1 (TRAF2) (300-305). TRAF2 in turn can recruit TRAF1 to the complex (301,302). RIP is necessary for the activation of NF-κB while FADD is important in induction of apoptosis. Activation of cytoplasmic proteins by RIP will result in phosphorylation of IκB allowing nuclear localization and activation of NF-κB (306). RAIDD has an N-terminal caspase recruitment domain (CARD), which mediates interaction with similar domains on caspase 2 (305). Using C-terminal death domain, RAIDD can interact with RIP and TRADD mediating the recruitment of caspase towards the oligomerized TNFR. TNFR1 needs TRADD as an intermediary between FADD and the receptor. FADD has a death effector domain (DED), which can interact with similar domains found at the N-terminus of caspase 8 (307). The multi-protein complex consisting of TNFRs, TRADD, FADD and caspase 8 is known as death inducing signaling complex (DISC) and it can induce the cleavage and activation of caspase 8 leading to the activation of downstream capases or the Bcl-2 family member BID (308-310). Ligation of TNF to its receptors can also lead to generation of reactive oxygen species in target cells, which can lead to apoptosis or necrosis (311). TNFR2 lacks a cytoplasmic death domain and has been shown to induce pathways of cell activation and survival. However, TNFR2 mediated cytotoxicity of target cells has been demonstrated (312). It has been proposed that the ligation of TNFR2 leads to the activation of 37 NF-κB and AP-1, which in turn will induce the transcriptional upregulation of membrane-bound TNF and FasL leading to apoptosis in an autotropic or paratropic manner (312). TRAF2 has been shown to signal for activation of JNK leading to phosphorylation of AP1 (313). Among the target genes upregulated by the TNF pathway, IAP proteins are found to play a major role in inhibiting downstream caspases (284). REGULATION OF CELL DEATH IN CYTOTOXIC CELLS Apoptotic cell death pathways play a critical role in shaping the repertoire of mature lymphocytes, which is crucial for the proper functioning of the immune system. Lymphocytes with developmental defects are removed early in lymphocyte development. During the peripheral immune response, activated lymphocytes have to be deleted after antigen clearance to assure that the highly active cytotoxic cells do not pose a threat to the host. Lymphocytes that suffer irreparable DNA damage, or those which are neglected by removal from necessary survival factors, undergo programmed cell death. The combined effects of all these events lead to the establishment of homeostasis (314). Peripheral T cells undergo apoptosis following high-intensity TCR signaling or absence of growth factors like IL-2 (315,316). Activated T cells downregulate Fas inhibitor FLIP and upregulate FasL to become more sensitive to cell death though Fas-FasL interactions (317,318). The activation-induced cell death (AICD) in T cells is induced by FasL or TNF, either selfinflicted or killed by adjacent cells (319,320). T cells transiently express both TNF and FasL during activation which can engage the corresponding receptors on themselves or on neighboring cells. Cell death due to Fas-FasL interaction is shown to be much faster than that is induced by TNF-TNFR2 (278). The reactive oxygen species pathway also has been implicated in T cell 38 AICD (321). Activation-induced NK cell death triggered by CD2, CD16 or CD94 was shown to be extremely rapid compared to AICD in T cells (322,323). Moreover, the AICD in NK cells were not inhibited by addition of antibodies against FasL or TNF. Granzyme B, which is leaked out of NK cells during activation, was found to induce their death by cleaving BID, resulting in mitochondrial tethering and apoptosis (324). NK cells express the protease inhibitor-9, which make complexes with granzymes. However, the ratio between granzyme and the inhibitor determines the outcome (324). It has been reported previously that NK cells recycle 3-4 times during an in vitro cytotoxicity assay enabling each cell to kill multiple targets (325). It can be hypothesized that various pathways act together to counteract the signals for AICD in these cells upon the activation. Several costimulatory signals have been demonstrated to save the T cells from AICD. Interaction of CD28 with CD80 (B7.1) or CD86 (B7.2) has been shown to protect T cells (326). Stimulation though CD28 was shown to enhance cytokine production, upregulate Bcl-xL and FLIP and downregulate expression of FasL (326-329). Inhibitor of apoptosis proteins (IAP) including hILP/XIAP, cIAP-1, cIAP-2, NAIP, survivin and Bruce are characterized by the presence of a 70 amino acid BIR domain, which shares similarity with similar domains in IAP proteins found in baculovirus (283,330). The role of hILP in regulation of T cell apoptosis has been demonstrated (331). hILP, cIAP-1 and cIAP-2 can bind and inactivate the active forms of caspase 3 and 7, but not the pro-enzymes (332-334). In addition, these proteins can bind, not only to the active caspase 9, but also to the procaspase 9, preventing its processing and activation (332). In response to strong apoptosis stimuli, cellular hILP can be cleaved by caspases leading to neutralization of its inhibitory effects on other caspases and apoptotic death (331). The cleavage products of hILP remain associated with 39 caspases and can be detected in peripheral blood T lymphocytes isolated from healthy individuals, which are stimulated to undergo AICD (335). It has been reported that IAPs could be regulated during exposure to TNF alpha (336,337). IAPs regulate TNF-mediated cytochrome c release and loss of mitochondrial membrane potential by inhibiting the signaling pathways upstream of mitochondrial events and apical caspase 8 (338). Members of the IAP family contain a C-terminal zinc-binding RING finger motif that is commonly found in ubiquitin ligases (339). RING finger containing IAP proteins were found to use their ubiquitinating properties to subject caspases to proteosomal degradation (340,341). Several caspase-independent survival mechanisms of IAP proteins have been documented in different tissues (342). Cellular apoptosis susceptibility protein (CAS) was identified as a homolog of yeast chromosome segregation protein (CSE1) (343-345), and it has been shown to be involved in both apoptosis and cell proliferation (346). CAS functions as a nuclear transport factor and mediates nuclear-to-cytosolic recycling of importin-alpha and probably other factors, which are important in cell proliferation and apoptosis (347). CAS expression is upregulated in proliferating cells while expression levels are low in quiescent cells and tissues (348,349). CAS contains a putative recognition site for MEK-1 phosphorylation at its N-terminus, indicating that its functions are regulated by phosphorylation (343,348). MEK plays an important role in proliferation as well as in TNF mediated apoptosis (350). Recent studies have demonstrated the regulation of apoptosis by proteins believed to be involved in totally different cellular functions. For example, the alpha-chain of the nascent polypeptide-associated complex has been found to bind and regulate the functions of FADD 40 (351). Inhibition of alpha NAC using anti-sense oligodeoxynucleotide resulted in an increased cell proliferation of CD8+ CTLs (352). In teleosts, several genes with homology to mammalian apoptosis regulators have been identified by analyzing the expressed sequence tags (EST) from zebrafish (353). Important among those are members of the Bcl-2 family, caspases, IAPs, DD containing receptors and death ligands (353). However, the biological significance of these findings can be known only after correlating the expression pattern to various tissues and cell types. Using cross-reacting antibodies, expression of apoptosis regulatory proteins have been identified in activated NCC, which might play an important role in protecting these cells from AICD (354,355). It has previously been demonstrated, that NCC lack the capacity to recycle (356). Thus, the role played by apoptosis regulatory proteins can be expected to be important for NCC functions (355). REFERENCES 1. Govaerts, A. 1960. Cellular Antibodies in Kidney Homotransplantation. Journal of Immunology 85:516. 2. Moller, E. 1965. Antagonistic Effects of Humoral Isoantibodies on in Vitro Cytotoxicity of Immune Lymphoid Cells. Journal of Experimental Medicine 122:11. 3. Wilson, D. B. 1965. Quantitative Studies on Behavior of Sensitized Lymphocytes in Vitro .I. Relationship to Degree of Destruction of Homologous Target Cells to Number of Lymphocytes and to Time of Contact in Culture and Consideration of Effects of Isimmune Serum. Journal of Experimental Medicine 122:143. 4. Wilson, D. B. 1965. Quantitative Studies on Behavior of Sensitized Lymphocytes in Vitro .2. Inhibitory Influence of Immune Suppressor Imuran on Destructive Reaction of Sensitized Lymphoid Cells Against Homologous Target Cells. Journal of Experimental Medicine 122:167. 5. Cerottin.JC, A. A. Nordin, and K. T. Brunner. 1970. Specific In-Vitro Cytotoxicity of Thymus-Derived Lymphocytes Sensitized to Alloantigens. Nature 228:1308. 41 6. Golstein, P., E. A. J. Svedmyr, H. Wigzell, and H. Blomgren. 1972. Cells Mediating Specific In-Vitro Cytotoxicity .2. Probable Autonomy of Thymus-Processed Lymphocytes (T Cells) for Killing of Allogeneic Target Cells. Journal of Experimental Medicine 135:890. 7. Waterhouse, N. J., C. J. P. Clarke, K. A. Sedelies, M. W. Teng, and J. A. Trapani. 2004. Cytotoxic lymphocytes; instigators of dramatic target cell death. Biochemical Pharmacology 68:1033. 8. Lieberman, J. 2003. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nat. Rev. Immunol. 3:361. 9. Doherty, P. C., and J. P. Christensen. 2000. Accessing complexity: The dynamics of virus-specific T cell responses. Annual Review of Immunology 18:561. 10. Russell, J. H., and T. J. Ley. 2002. Lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 20:323. 11. Russell, J. H., and C. B. Dobos. 1980. Mechanisms of immune lysis. II. CTL-induced nuclear disintegration of the target begins within minutes of cell contact. J. Immunol. 125:1256. 12. Russell, J. H., V. R. Masakowski, and C. B. Dobos. 1980. Mechanisms of immune lysis. I. Physiological distinction between target cell death mediated by cytotoxic T lymphocytes and antibody plus complement. J. Immunol. 124:1100. 13. Sellins, K. S., and J. J. Cohen. 1991. Cytotoxic T lymphocytes induce different types of DNA damage in target cells of different origins. J. Immunol. 147:795. 14. Pardo, J., S. Balkow, A. Anel, and M. M. Simon. 2002. The differential contribution of granzyme A and granzyme B in cytotoxic T lymphocyte-mediated apoptosis is determined by the quality of target cells. Eur. J. Immunol. 32:1980. 15. Graves, S. S., D. L. Evans, and D. L. Dawe. 1985. Antiprotozoan activity of nonspecific cytotoxic cells (NCC) from the channel catfish (Ictalurus punctatus). J. Immunol. 134:78. 16. Evans, D. L., L. Jaso-Friedmann, E. E. Smith, Jr., J. A. St, H. S. Koren, and D. T. Harris. 1988. Identification of a putative antigen receptor on fish nonspecific cytotoxic cells with monoclonal antibodies. J. Immunol. 141:324. 17. Jaso-Friedmann, L., D. T. Harris, J. A. St, H. S. Koren, and D. L. Evans. 1990. A monoclonal antibody-purified soluble target cell antigen inhibits nonspecific cytotoxic cell activity. J. Immunol. 144:2413. 18. Evans, D. L., J. H. Leary, III, and L. Jaso-Friedmann. 1998. Nonspecific cytotoxic cell receptor protein-1: a novel (predicted) type III membrane receptor on the teleost equivalent of natural killer cells recognizes conventional antigen. Cell Immunol. 187:19. 42 19. Jaso-Friedmann, L., D. S. Peterson, D. S. Gonzalez, and D. L. Evans. 2002. The antigen receptor (NCCRP-1) on catfish and zebrafish nonspecific cytotoxic cells belongs to a new gene family characterized by an F-box-associated domain. J. Mol. Evol. 54:386. 20. Jaso-Friedmann, L., and D. L. Evans. 1999. Mechanisms of cellular cytotoxic innate resistance in tilapia (Oreochromis nilotica). Dev. Comp Immunol. 23:27. 21. Faisal, M., I. I. Ahmed, G. Peters, and E. L. Cooper. 1989. Natural Cytotoxicity of Tilapia Leukocytes. Diseases of Aquatic Organisms 7:17. 22. McKinney, E. C., and M. C. Schmale. 1994. Damselfish with neurofibromatosis exhibit cytotoxicity toward tumor targets. Dev. Comp Immunol. 18:305. 23. Suzumura, E., O. Kurata, N. Okamoto, and Y. Ikeda. 1994. Characteristics of Natural Killer-Like Cells in Carp. Fish Pathology 29:199. 24. Shen, L., T. B. Stuge, E. Bengten, M. Wilson, V. G. Chinchar, J. P. Naftel, J. M. Bernanke, L. W. Clem, and N. W. Miller. 2004. Identification and characterization of clonal NK-like cells from channel catfish (Ictalurus punctatus). Dev. Comp Immunol. 28:139. 25. Shen, L., T. B. Stuge, H. Zhou, M. Khayat, K. S. Barker, S. M. Quiniou, M. Wilson, E. Bengten, V. G. Chinchar, L. W. Clem, and N. W. Miller. 2002. Channel catfish cytotoxic cells: a mini-review. Dev. Comp Immunol. 26:141. 26. Bishop, G. R., L. Jaso-Friedmann, and D. L. Evans. 2000. Activation-induced programmed cell death of nonspecific cytotoxic cells and inhibition by apoptosis regulatory factors. Cell Immunol. 199:126. 27. Jaso-Friedmann, L., J. H. Leary, III, and D. L. Evans. 2000. Role of nonspecific cytotoxic cells in the induction of programmed cell death of pathogenic protozoans: participation of the Fas ligand-Fas receptor system. Exp. Parasitol. 96:75. 28. Long, S., M. Wilson, E. Bengten, L. W. Clem, N. W. Miller, and V. G. Chinchar. 2004. Identification and characterization of a FasL-like protein and cDNAs encoding the channel catfish death-inducing signaling complex. Immunogenetics 56:518. 29. Greenlee, A. R., R. A. Brown, and S. S. Ristow. 1991. Nonspecific cytotoxic cells of rainbow trout (Oncorhynchus mykiss) kill YAC-1 targets by both necrotic and apoptic mechanisms. Dev. Comp Immunol. 15:153. 30. Carlson, R. L., D. L. Evans, and S. S. Graves. 1985. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). V. Metabolic requirements of lysis. Dev. Comp Immunol. 9:271. 31. Zhou, H., T. B. Stuge, N. W. Miller, E. Bengten, J. P. Naftel, J. M. Bernanke, V. G. Chinchar, L. W. Clem, and M. Wilson. 2001. Heterogeneity of channel catfish CTL with respect to target recognition and cytotoxic mechanisms employed. J. Immunol. 167:1325. 43 32. Jaso-Friedmann, L., J. H. Leary, III, Z. Weisman, and D. L. Evans. 1996. Activation of nonspecific cytotoxic cells with a multiple antigenic peptide: specificity and requirements for receptor crosslinkage. Cell Immunol. 170:195. 33. Suda, T., T. Takahashi, P. Golstein, and S. Nagata. 1993. Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 75:1169. 34. Griffith, T. S., T. Brunner, S. M. Fletcher, D. R. Green, and T. A. Ferguson. 1995. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270:1189. 35. Bossi, G., and G. M. Griffiths. 1999. Degranulation plays an essential part in regulating cell surface expression of Fas ligand in T cells and natural killer cells. Nat. Med. 5:90. 36. Davis, D. M., I. Chiu, M. Fassett, G. B. Cohen, O. Mandelboim, and J. L. Strominger. 1999. The human natural killer cell immune synapse. Proc. Natl. Acad. Sci. U. S. A 96:15062. 37. Davis, D. M. 2002. Assembly of the immunological synapse for T cells and NK cells. Trends Immunol. 23:356. 38. Burkhardt, J. K., S. Hester, C. K. Lapham, and Y. Argon. 1990. The lytic granules of natural killer cells are dual-function organelles combining secretory and pre-lysosomal compartments. J. Cell Biol. 111:2327. 39. Peters, P. J., J. Borst, V. Oorschot, M. Fukuda, O. Krahenbuhl, J. Tschopp, J. W. Slot, and H. J. Geuze. 1991. Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J. Exp. Med. 173:1099. 40. Masson, D., P. J. Peters, H. J. Geuze, J. Borst, and J. Tschopp. 1990. Interaction of chondroitin sulfate with perforin and granzymes of cytolytic T-cells is dependent on pH. Biochemistry 29:11229. 41. Clark, R., and G. M. Griffiths. 2003. Lytic granules, secretory lysosomes and disease. Curr. Opin. Immunol. 15:516. 42. Simon, M. M., M. D. Kramer, M. Prester, and S. Gay. 1991. Mouse T-cell associated serine proteinase 1 degrades collagen type IV: a structural basis for the migration of lymphocytes through vascular basement membranes. Immunology 73:117. 43. Trapani, J. A. 2001. Granzymes: a family of lymphocyte granule serine proteases. Genome Biol. 2: Reviews 3014. 44. Fraser, S. A., R. Karimi, M. Michalak, and D. Hudig. 2000. Perforin lytic activity is controlled by calreticulin. J. Immunol. 164:4150. 44 45. Greenberg, A. H., N. Khalil, B. Pohajdak, M. Talgoy, P. Henkart, and F. W. Orr. 1986. NK-leukocyte chemotactic factor (NK-LCF): a large granular lymphocyte (LGL) granule-associated chemotactic factor. J. Immunol. 137:3224. 46. Wagner, L., O. O. Yang, E. A. Garcia-Zepeda, Y. Ge, S. A. Kalams, B. D. Walker, M. S. Pasternack, and A. D. Luster. 1998. Beta-chemokines are released from HIV-1-specific cytolytic T-cell granules complexed to proteoglycans. Nature 391:908. 47. Stevens, R. L., M. M. Kamada, and W. E. Serafin. 1989. Structure and function of the family of proteoglycans that reside in the secretory granules of natural killer cells and other effector cells of the immune response. Curr. Top. Microbiol. Immunol. 140:93. 48. Stenger, S., D. A. Hanson, R. Teitelbaum, P. Dewan, K. R. Niazi, C. J. Froelich, T. Ganz, S. Thoma-Uszynski, A. Melian, C. Bogdan, S. A. Porcelli, B. R. Bloom, A. M. Krensky, and R. L. Modlin. 1998. An antimicrobial activity of cytolytic T cells mediated by granulysin. Science 282:121. 49. Chuang, E., M. L. Alegre, C. S. Duckett, P. J. Noel, M. G. Vander Heiden, and C. B. Thompson. 1997. Interaction of CTLA-4 with the clathrin-associated protein AP50 results in ligand-independent endocytosis that limits cell surface expression. J. Immunol. 159:144. 50. Hink-Schauer, C., E. Estebanez-Perpina, F. C. Kurschus, W. Bode, and D. E. Jenne. 2003. Crystal structure of the apoptosis-inducing human granzyme A dimer. Nat. Struct. Biol. 10:535. 51. Smyth, M. J., M. D. O' Connor, and J. A. Trapani. 1996. Granzymes: a variety of serine protease specificities encoded by genetically distinct subfamilies. J. Leukoc. Biol. 60:555. 52. Grossman, W. J., P. A. Revell, Z. H. Lu, H. Johnson, A. J. Bredemeyer, and T. J. Ley. 2003. The orphan granzymes of humans and mice. Curr. Opin. Immunol. 15:544. 53. Estebanez-Perpina, E., P. Fuentes-Prior, D. Belorgey, M. Braun, R. Kiefersauer, K. Maskos, R. Huber, H. Rubin, and W. Bode. 2000. Crystal structure of the caspase activator human granzyme B, a proteinase highly specific for an Asp-P1 residue. Biol. Chem. 381:1203. 54. Hink-Schauer, C., E. Estebanez-Perpina, E. Wilharm, P. Fuentes-Prior, W. Klinkert, W. Bode, and D. E. Jenne. 2002. The 2.2-A crystal structure of human pro-granzyme K reveals a rigid zymogen with unusual features. J. Biol. Chem. 277:50923. 55. Sattar, R., S. A. Ali, and A. Abbasi. 2003. Bioinformatics of granzymes: sequence comparison and structural studies on granzyme family by homology modeling. Biochem. Biophys. Res. Commun. 308:726. 56. McGuire, M. J., P. E. Lipsky, and D. L. Thiele. 1993. Generation of active myeloid and lymphoid granule serine proteases requires processing by the granule thiol protease dipeptidyl peptidase I. J. Biol. Chem. 268:2458. 45 57. Griffiths, G. M., and S. Isaaz. 1993. Granzymes A and B are targeted to the lytic granules of lymphocytes by the mannose-6-phosphate receptor. J. Cell Biol. 120:885. 58. Wilharm, E., M. A. Parry, R. Friebel, H. Tschesche, G. Matschiner, C. P. Sommerhoff, and D. E. Jenne. 1999. Generation of catalytically active granzyme K from Escherichia coli inclusion bodies and identification of efficient granzyme K inhibitors in human plasma. J. Biol. Chem. 274:27331. 59. Bell, J. K., D. H. Goetz, S. Mahrus, J. L. Harris, R. J. Fletterick, and C. S. Craik. 2003. The oligomeric structure of human granzyme A is a determinant of its extended substrate specificity. Nat. Struct. Biol. 10:527. 60. Bleackley, R. C., C. G. Lobe, B. Duggan, N. Ehrman, C. Fregeau, M. Meier, M. Letellier, C. Havele, J. Shaw, and V. Paetkau. 1988. The isolation and characterization of a family of serine protease genes expressed in activated cytotoxic T lymphocytes. Immunol. Rev. 103:5. 61. Pham, C. T., D. M. MacIvor, B. A. Hug, J. W. Heusel, and T. J. Ley. 1996. Long-range disruption of gene expression by a selectable marker cassette. Proc. Natl. Acad. Sci. U. S. A 93:13090. 62. Garcia-Sanz, J. A., H. R. MacDonald, D. E. Jenne, J. Tschopp, and M. Nabholz. 1990. Cell specificity of granzyme gene expression. J. Immunol. 145:3111. 63. Kelso, A., E. O. Costelloe, B. J. Johnson, P. Groves, K. Buttigieg, and D. R. Fitzpatrick. 2002. The genes for perforin, granzymes A-C and IFN-gamma are differentially expressed in single CD8(+) T cells during primary activation. Int. Immunol. 14:605. 64. Sasson, R., A. Dantes, K. Tajima, and A. Amsterdam. 2003. Novel genes modulated by FSH in normal and immortalized FSH-responsive cells: new insights into the mechanism of FSH action. FASEB J. 17:1256. 65. Hu, S. X., S. Wang, J. P. Wang, G. B. Mills, Y. Zhou, and H. J. Xu. 2003. Expression of endogenous granzyme B in a subset of human primary breast carcinomas. Br. J. Cancer 89:135. 66. Hirst, C. E., M. S. Buzza, V. R. Sutton, J. A. Trapani, K. L. Loveland, and P. I. Bird. 2001. Perforin-independent expression of granzyme B and proteinase inhibitor 9 in human testis and placenta suggests a role for granzyme B-mediated proteolysis in reproduction. Mol. Hum. Reprod. 7:1133. 67. Spaeny-Dekking, E. H., W. L. Hanna, A. M. Wolbink, P. C. Wever, A. J. Kummer, A. J. Swaak, J. M. Middeldorp, H. G. Huisman, C. J. Froelich, and C. E. Hack. 1998. Extracellular granzymes A and B in humans: detection of native species during CTL responses in vitro and in vivo. J. Immunol. 160:3610. 68. Hermsen, C. C., Y. Konijnenberg, L. Mulder, C. Loe, D. M. van, J. W. van der Meer, G. J. van Mierlo, W. M. Eling, C. E. Hack, and R. W. Sauerwein. 2003. Circulating 46 concentrations of soluble granzyme A and B increase during natural and experimental Plasmodium falciparum infections. Clin. Exp. Immunol. 132:467. 69. Tak, P. P., L. Spaeny-Dekking, M. C. Kraan, F. C. Breedveld, C. J. Froelich, and C. E. Hack. 1999. The levels of soluble granzyme A and B are elevated in plasma and synovial fluid of patients with rheumatoid arthritis (RA). Clin. Exp. Immunol. 116:366. 70. Suemoto, T., M. Taniguchi, S. Shiosaka, and S. Yoshida. 1999. cDNA cloning and expression of a novel serine protease in the mouse brain. Brain Res. Mol. Brain Res. 70:273. 71. Taniguchi, M., N. Tani, T. Suemoto, I. Ishimoto, S. Shiosaka, and S. Yoshida. 1999. High expression of alternative transcript of granzyme M in the mouse retina. Neurosci. Res. 34:115. 72. Trambas, C. M., and G. M. Griffiths. 2003. Delivering the kiss of death. Nature Immunology 4:399. 73. Abrams, S. I., and Z. Brahmi. 1986. The functional loss of human natural killer cell activity induced by K562 is reversible via an interleukin-2-dependent mechanism. Cell Immunol. 101:558. 74. Doherty, P. C. 1993. Cell-mediated cytotoxicity. Cell 75:607. 75. Podack, E. R., and A. Kupfer. 1991. T-cell effector functions: mechanisms for delivery of cytotoxicity and help. Annu. Rev. Cell Biol. 7:479. 76. Peters, P. J., H. J. Geuze, H. A. van der Donk, and J. Borst. 1990. A new model for lethal hit delivery by cytotoxic T lymphocytes. Immunol. Today 11:28. 77. Geiger, B., D. Rosen, and G. Berke. 1982. Spatial relationships of microtubuleorganizing centers and the contact area of cytotoxic T lymphocytes and target cells. J. Cell Biol. 95:137. 78. Kupfer, A., and G. Dennert. 1984. Reorientation of the microtubule-organizing center and the Golgi apparatus in cloned cytotoxic lymphocytes triggered by binding to lysable target cells. J. Immunol. 133:2762. 79. Kupfer, A., G. Dennert, and S. J. Singer. 1985. The reorientation of the Golgi apparatus and the microtubule-organizing center in the cytotoxic effector cell is a prerequisite in the lysis of bound target cells. J. Mol. Cell Immunol. 2:37. 80. van der Merwe, P. A. 2002. Formation and function of the immunological synapse. Curr. Opin. Immunol. 14:293. 81. Djeu, J. Y., K. Jiang, and S. Wei. 2002. A view to a kill: signals triggering cytotoxicity. Clin. Cancer Res. 8:636. 47 82. Brumbaugh, K. M., B. A. Binstadt, D. D. Billadeau, R. A. Schoon, C. J. Dick, R. M. Ten, and P. J. Leibson. 1997. Functional role for Syk tyrosine kinase in natural killer cellmediated natural cytotoxicity. J. Exp. Med. 186:1965. 83. Billadeau, D. D., K. M. Brumbaugh, C. J. Dick, R. A. Schoon, X. R. Bustelo, and P. J. Leibson. 1998. The Vav-Rac1 pathway in cytotoxic lymphocytes regulates the generation of cell-mediated killing. J. Exp. Med. 188:549. 84. Galandrini, R., G. Palmieri, M. Piccoli, L. Frati, and A. Santoni. 1999. Role for the Rac1 exchange factor Vav in the signaling pathways leading to NK cell cytotoxicity. J. Immunol. 162:3148. 85. Wei, S., A. M. Gamero, J. H. Liu, A. A. Daulton, N. I. Valkov, J. A. Trapani, A. C. Larner, M. J. Weber, and J. Y. Djeu. 1998. Control of lytic function by mitogen-activated protein kinase/extracellular regulatory kinase 2 (ERK2) in a human natural killer cell line: identification of perforin and granzyme B mobilization by functional ERK2. J. Exp. Med. 187:1753. 86. Trotta, R., K. A. Puorro, M. Paroli, L. Azzoni, B. Abebe, L. C. Eisenlohr, and B. Perussia. 1998. Dependence of both spontaneous and antibody-dependent, granule exocytosis-mediated NK cell cytotoxicity on extracellular signal-regulated kinases. J. Immunol. 161:6648. 87. Carretero, M., M. Llano, F. Navarro, T. Bellon, and M. Lopez-Botet. 2000. Mitogenactivated protein kinase activity is involved in effector functions triggered by the CD94/NKG2-C NK receptor specific for HLA-E. Eur. J. Immunol. 30:2842. 88. Khurana, D., and P. J. Leibson. 2003. Regulation of lymphocyte-mediated killing by GTP-binding proteins. J. Leukoc. Biol. 73:333. 89. Young, J. D., H. Hengartner, E. R. Podack, and Z. A. Cohn. 1986. Purification and characterization of a cytolytic pore-forming protein from granules of cloned lymphocytes with natural killer activity. Cell 44:849. 90. Browne, K. A., E. Blink, V. R. Sutton, C. J. Froelich, D. A. Jans, and J. A. Trapani. 1999. Cytosolic delivery of granzyme B by bacterial toxins: evidence that endosomal disruption, in addition to transmembrane pore formation, is an important function of perforin. Mol. Cell Biol. 19:8604. 91. Froelich, C. J., K. Orth, J. Turbov, P. Seth, R. Gottlieb, B. Babior, G. M. Shah, R. C. Bleackley, V. M. Dixit, and W. Hanna. 1996. New paradigm for lymphocyte granulemediated cytotoxicity. Target cells bind and internalize granzyme B, but an endosomolytic agent is necessary for cytosolic delivery and subsequent apoptosis. J. Biol. Chem. 271:29073. 92. Shi, L., S. Mai, S. Israels, K. Browne, J. A. Trapani, and A. H. Greenberg. 1997. Granzyme B (GraB) autonomously crosses the cell membrane and perforin initiates apoptosis and GraB nuclear localization. J. Exp. Med. 185:855. 48 93. Pinkoski, M. J., M. Hobman, J. A. Heibein, K. Tomaselli, F. Li, P. Seth, C. J. Froelich, and R. C. Bleackley. 1998. Entry and trafficking of granzyme B in target cells during granzyme B-perforin-mediated apoptosis. Blood 92:1044. 94. Motyka, B., G. Korbutt, M. J. Pinkoski, J. A. Heibein, A. Caputo, M. Hobman, M. Barry, I. Shostak, T. Sawchuk, C. F. Holmes, J. Gauldie, and R. C. Bleackley. 2000. Mannose 6phosphate/insulin-like growth factor II receptor is a death receptor for granzyme B during cytotoxic T cell-induced apoptosis. Cell 103:491. 95. Trapani, J. A., V. R. Sutton, K. Y. Thia, Y. Q. Li, C. J. Froelich, D. A. Jans, M. S. Sandrin, and K. A. Browne. 2003. A clathrin/dynamin- and mannose-6-phosphate receptor-independent pathway for granzyme B-induced cell death. J. Cell Biol. 160:223. 96. Dressel, R., S. M. Raja, S. Honing, T. Seidler, C. J. Froelich, F. K. Von, and E. Gunther. 2004. Granzyme-mediated Cytotoxicity Does Not Involve the Mannose 6-Phosphate Receptors on Target Cells. J. Biol. Chem. 279:20200. 97. Kurschus, F. C., R. Bruno, E. Fellows, C. S. Falk, and D. E. Jenne. 2004. Membrane receptors are not required to deliver granzyme B during killer cell attack. Blood. 98. Galvin, J. P., L. H. Spaeny-Dekking, B. Wang, P. Seth, C. E. Hack, and C. J. Froelich. 1999. Apoptosis induced by granzyme B-glycosaminoglycan complexes: implications for granule-mediated apoptosis in vivo. J. Immunol. 162:5345. 99. Metkar, S. S., B. Wang, M. guilar-Santelises, S. M. Raja, L. Uhlin-Hansen, E. Podack, J. A. Trapani, and C. J. Froelich. 2002. Cytotoxic cell granule-mediated apoptosis: perforin delivers granzyme B-serglycin complexes into target cells without plasma membrane pore formation. Immunity 16:417. 100. Raja, S. M., B. Wang, M. Dantuluri, U. R. Desai, B. Demeler, K. Spiegel, S. S. Metkar, and C. J. Froelich. 2002. Cytotoxic cell granule-mediated apoptosis. Characterization of the macromolecular complex of granzyme B with serglycin. J. Biol. Chem. 277:49523. 101. Waugh, S. M., J. L. Harris, R. Fletterick, and C. S. Craik. 2000. The structure of the proapoptotic protease granzyme B reveals the molecular determinants of its specificity. Nat. Struct. Biol. 7:762. 102. Matsumoto, R., A. Sali, N. Ghildyal, M. Karplus, and R. L. Stevens. 1995. Packaging of proteases and proteoglycans in the granules of mast cells and other hematopoietic cells. A cluster of histidines on mouse mast cell protease 7 regulates its binding to heparin serglycin proteoglycans. J. Biol. Chem. 270:19524. 103. Veugelers, K., B. Motyka, C. Frantz, I. Shostak, T. Sawchuk, and R. C. Bleackley. 2004. The granzyme B-serglycin complex from cytotoxic granules requires dynamin for endocytosis. Blood. 49 104. Gross, C., W. Koelch, A. DeMaio, N. Arispe, and G. Multhoff. 2003. Cell surface-bound heat shock protein 70 (Hsp70) mediates perforin-independent apoptosis by specific binding and uptake of granzyme B. J. Biol. Chem. 278:41173. 105. Henkart, P. A. 1985. Mechanism of lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 3:31. 106. Uellner, R., M. J. Zvelebil, J. Hopkins, J. Jones, L. K. MacDougall, B. P. Morgan, E. Podack, M. D. Waterfield, and G. M. Griffiths. 1997. Perforin is activated by a proteolytic cleavage during biosynthesis which reveals a phospholipid-binding C2 domain. EMBO J. 16:7287. 107. Young, J. D., Z. A. Cohn, and E. R. Podack. 1986. The ninth component of complement and the pore-forming protein (perforin 1) from cytotoxic T cells: structural, immunological, and functional similarities. Science 233:184. 108. Podack, E. R., and G. Dennert. 1983. Assembly of two types of tubules with putative cytolytic function by cloned natural killer cells. Nature 302:442. 109. Tschopp, J., D. Masson, and K. K. Stanley. 1986. Structural/functional similarity between proteins involved in complement- and cytotoxic T-lymphocyte-mediated cytolysis. Nature 322:831. 110. Kagi, D., B. Ledermann, K. Burki, P. Seiler, B. Odermatt, K. J. Olsen, E. R. Podack, R. M. Zinkernagel, and H. Hengartner. 1994. Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice. Nature 369:31. 111. Mullbacher, A., K. Ebnet, R. V. Blanden, R. T. Hla, T. Stehle, C. Museteanu, and M. M. Simon. 1996. Granzyme A is critical for recovery of mice from infection with the natural cytopathic viral pathogen, ectromelia. Proc. Natl. Acad. Sci. U. S. A 93:5783. 112. Walsh, C. M., M. Matloubian, C. C. Liu, R. Ueda, C. G. Kurahara, J. L. Christensen, M. T. Huang, J. D. Young, R. Ahmed, and W. R. Clark. 1994. Immune function in mice lacking the perforin gene. Proc. Natl. Acad. Sci. U. S. A 91:10854. 113. Kojima, H., N. Shinohara, S. Hanaoka, Y. Someya-Shirota, Y. Takagaki, H. Ohno, T. Saito, T. Katayama, H. Yagita, K. Okumura, and . 1994. Two distinct pathways of specific killing revealed by perforin mutant cytotoxic T lymphocytes. Immunity 1:357. 114. Duke, R. C., R. Chervenak, and J. J. Cohen. 1983. Endogenous endonuclease-induced DNA fragmentation: an early event in cell-mediated cytolysis. Proc. Natl. Acad. Sci. U. S. A 80:6361. 115. Matter, A. 1979. Microcinematographic and electron microscopic analysis of target cell lysis induced by cytotoxic T lymphocytes. Immunology 36:179. 116. Russell, J. H. 1983. Internal disintegration model of cytotoxic lymphocyte-induced target damage. Immunol. Rev. 72:97. 50 117. Shiver, J. W., L. Su, and P. A. Henkart. 1992. Cytotoxicity with target DNA breakdown by rat basophilic leukemia cells expressing both cytolysin and granzyme A. Cell 71:315. 118. Bashford, C. L., G. Menestrina, P. A. Henkart, and C. A. Pasternak. 1988. Cell damage by cytolysin. Spontaneous recovery and reversible inhibition by divalent cations. J. Immunol. 141:3965. 119. Chang, T. W., and H. N. Eisen. 1980. Effects of N alpha-tosyl-L-lysylchloromethylketone on the activity of cytotoxic T lymphocytes. J. Immunol. 124:1028. 120. Quan, P. C., T. Ishizaka, and B. R. Bloom. 1982. Studies on the mechanism of NK cell lysis. J. Immunol. 128:1786. 121. Redelman, D., and D. Hudig. 1980. The mechanism of cell-mediated cytotoxicity. I. Killing by murine cytotoxic T lymphocytes requires cell surface thiols and activated proteases. J. Immunol. 124:870. 122. Redelman, D., and D. Hudig. 1983. The mechanism of cell-mediated cytotoxicity. III. Protease-specific inhibitors preferentially block later events in cytotoxic T lymphocytemediated lysis than do inhibitors of methylation or thiol-reactive agents. Cell Immunol. 81:9. 123. Hudig, D., T. Haverty, C. Fulcher, D. Redelman, and J. Mendelsohn. 1981. Inhibition of human natural cytotoxicity by macromolecular antiproteases. J. Immunol. 126:1569. 124. Hudig, D., D. Redelman, and L. L. Minning. 1984. The requirement for proteinase activity for human lymphocyte-mediated natural cytotoxicity (NK): evidence that the proteinase is serine dependent and has aromatic amino acid specificity of cleavage. J. Immunol. 133:2647. 125. Lavie, G., Z. Leib, and C. Servadio. 1985. The mechanism of human NK cell-mediated cytotoxicity. Mode of action of surface-associated proteases in the early stages of the lytic reaction. J. Immunol. 135:1470. 126. Hatcher, V. B., M. S. Oberman, G. S. Lazarus, and A. I. Grayzel. 1978. A cytotoxic proteinase isolated from human lymphocytes. J. Immunol. 120:665. 127. Darmon, A. J., D. W. Nicholson, and R. C. Bleackley. 1995. Activation of the apoptotic protease CPP32 by cytotoxic T-cell-derived granzyme B. Nature 377:446. 128. Adrain, C., B. M. Murphy, and S. J. Martin. 2004. Molecular ordering of the caspase activation cascade initiated by the CTL/NK protease granzyme B. J. Biol. Chem. 129. Sarin, A., M. S. Williams, M. A. exander-Miller, J. A. Berzofsky, C. M. Zacharchuk, and P. A. Henkart. 1997. Target cell lysis by CTL granule exocytosis is independent of ICE/Ced-3 family proteases. Immunity 6:209. 51 130. Trapani, J. A., D. A. Jans, P. J. Jans, M. J. Smyth, K. A. Browne, and V. R. Sutton. 1998. Efficient nuclear targeting of granzyme B and the nuclear consequences of apoptosis induced by granzyme B and perforin are caspase-dependent, but cell death is caspaseindependent. J. Biol. Chem. 273:27934. 131. Sharif-Askari, E., A. Alam, E. Rheaume, P. J. Beresford, C. Scotto, K. Sharma, D. Lee, W. E. DeWolf, M. E. Nuttall, J. Lieberman, and R. P. Sekaly. 2001. Direct cleavage of the human DNA fragmentation factor-45 by granzyme B induces caspase-activated DNase release and DNA fragmentation. EMBO J. 20:3101. 132. Thomas, D. A., C. Du, M. Xu, X. Wang, and T. J. Ley. 2000. DFF45/ICAD can be directly processed by granzyme B during the induction of apoptosis. Immunity 12:621. 133. Wolf, B. B., M. Schuler, F. Echeverri, and D. R. Green. 1999. Caspase-3 is the primary activator of apoptotic DNA fragmentation via DNA fragmentation factor-45/inhibitor of caspase-activated DNase inactivation. J. Biol. Chem. 274:30651. 134. Zhang, D., P. J. Beresford, A. H. Greenberg, and J. Lieberman. 2001. Granzymes A and B directly cleave lamins and disrupt the nuclear lamina during granule-mediated cytolysis. Proc. Natl. Acad. Sci. U. S. A 98:5746. 135. Browne, K. A., R. W. Johnstone, D. A. Jans, and J. A. Trapani. 2000. Filamin (280-kDa actin-binding protein) is a caspase substrate and is also cleaved directly by the cytotoxic T lymphocyte protease granzyme B during apoptosis. J. Biol. Chem. 275:39262. 136. Kuwana, T., M. R. Mackey, G. Perkins, M. H. Ellisman, M. Latterich, R. Schneiter, D. R. Green, and D. D. Newmeyer. 2002. Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell 111:331. 137. Alimonti, J. B., L. F. Shi, P. K. Baijal, and A. H. Greenberg. 2001. Granzyme B induces bid-mediated cytochrome C release and mitochondrial permeability transition. ScientificWorldJournal. 1:49. 138. Heibein, J. A., I. S. Goping, M. Barry, M. J. Pinkoski, G. C. Shore, D. R. Green, and R. C. Bleackley. 2000. Granzyme B-mediated cytochrome c release is regulated by the Bcl2 family members bid and Bax. J. Exp. Med. 192:1391. 139. Heibein, J. A., M. Barry, B. Motyka, and R. C. Bleackley. 1999. Granzyme B-induced loss of mitochondrial inner membrane potential (Delta Psi m) and cytochrome c release are caspase independent. J. Immunol. 163:4683. 140. Sutton, V. R., J. E. Davis, M. Cancilla, R. W. Johnstone, A. A. Ruefli, K. Sedelies, K. A. Browne, and J. A. Trapani. 2000. Initiation of apoptosis by granzyme B requires direct cleavage of bid, but not direct granzyme B-mediated caspase activation. J. Exp. Med. 192:1403. 52 141. Thomas, D. A., L. Scorrano, G. V. Putcha, S. J. Korsmeyer, and T. J. Ley. 2001. Granzyme B can cause mitochondrial depolarization and cell death in the absence of BID, BAX, and BAK. Proc. Natl. Acad. Sci. U. S. A 98:14985. 142. MacDonald, G., L. Shi, V. C. Vande, J. Lieberman, and A. H. Greenberg. 1999. Mitochondria-dependent and -independent regulation of Granzyme B-induced apoptosis. J. Exp. Med. 189:131. 143. Wang, G. Q., E. Wieckowski, L. A. Goldstein, B. R. Gastman, A. Rabinovitz, A. Gambotto, S. Li, B. Fang, X. M. Yin, and H. Rabinowich. 2001. Resistance to granzyme B-mediated cytochrome c release in Bak-deficient cells. J. Exp. Med. 194:1325. 144. Han, J., L. A. Goldstein, B. R. Gastman, C. J. Froelich, X. M. Yin, and H. Rabinowich. 2004. Degradation of MCL-1 by granzyme B: Implications for bim-mediated mitochondrial apoptotic events. J. Biol. Chem. 145. Han, J., L. A. Goldstein, B. R. Gastman, A. Rabinovitz, and H. Rabinowich. 2005. Disruption of MCL-1/BIM complex in granzyme B-mediated mitochondrial apoptosis. J. Biol. Chem. 146. Andrade, F., S. Roy, D. Nicholson, N. Thornberry, A. Rosen, and L. Casciola-Rosen. 1998. Granzyme B directly and efficiently cleaves several downstream caspase substrates: implications for CTL-induced apoptosis. Immunity 8:451. 147. Froelich, C. J., W. L. Hanna, G. G. Poirier, P. J. Duriez, D. D' Amours, G. S. Salvesen, E. S. Alnemri, W. C. Earnshaw, and G. M. Shah. 1996. Granzyme B/perforin-mediated apoptosis of Jurkat cells results in cleavage of poly(ADP-ribose) polymerase to the 89kDa apoptotic fragment and less abundant 64-kDa fragment. Biochem. Biophys. Res. Commun. 227:658. 148. Sebbagh, M., J. Hamelin, J. Bertoglio, E. Solary, and J. Breard. 2005. Direct cleavage of ROCK II by granzyme B induces target cell membrane blebbing in a caspaseindependent manner. J. Exp. Med. 201:465. 149. Wieckowski, E., G. Q. Wang, B. R. Gastman, L. A. Goldstein, and H. Rabinowich. 2002. Granzyme B-mediated degradation of T-cell receptor zeta chain. Cancer Res. 62:4884. 150. Howe, A. Y., J. U. Jung, and R. C. Desrosiers. 1998. Zeta chain of the T-cell receptor interacts with nef of simian immunodeficiency virus and human immunodeficiency virus type 2. J. Virol. 72:9827. 151. Xu, X. N., B. Laffert, G. R. Screaton, M. Kraft, D. Wolf, W. Kolanus, J. Mongkolsapay, A. J. McMichael, and A. S. Baur. 1999. Induction of Fas ligand expression by HIV involves the interaction of Nef with the T cell receptor zeta chain. J. Exp. Med. 189:1489. 152. Nakajima, H., H. L. Park, and P. A. Henkart. 1995. Synergistic roles of granzymes A and B in mediating target cell death by rat basophilic leukemia mast cell tumors also expressing cytolysin/perforin. J. Exp. Med. 181:1037. 53 153. Beresford, P. J., Z. Xia, A. H. Greenberg, and J. Lieberman. 1999. Granzyme A loading induces rapid cytolysis and a novel form of DNA damage independently of caspase activation. Immunity 10:585. 154. Beresford, P. J., D. Zhang, D. Y. Oh, Z. Fan, E. L. Greer, M. L. Russo, M. Jaju, and J. Lieberman. 2001. Granzyme A activates an endoplasmic reticulum-associated caspaseindependent nuclease to induce single-stranded DNA nicks. J. Biol. Chem. 276:43285. 155. Fan, Z. S., P. J. Beresford, D. Y. Oh, D. Zhang, and J. Lieberman. 2003. Tumor suppressor NM23-H1 is a granzyme A-activated DNase during CTL-mediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell 112:659. 156. Fan, Z. S., P. J. Beresford, D. Zhang, and J. Lieberman. 2002. HMG2 interacts with the nucleosome assembly protein SET and is a target of the cytotoxic T-lymphocyte protease granzyme A. Molecular and Cellular Biology 22:2810. 157. Fan, Z. S., P. J. Beresford, D. Zhang, Z. Xu, C. D. Novina, A. Yoshida, Y. Pommier, and J. Lieberman. 2003. Cleaving the oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nature Immunology 4:145. 158. Fan, Z., P. J. Beresford, D. Y. Oh, D. Zhang, and J. Lieberman. 2003. Tumor suppressor NM23-H1 is a granzyme A-activated DNase during CTL-mediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell 112:659. 159. Ma, D., Z. Xing, B. Liu, N. G. Pedigo, S. G. Zimmer, Z. Bai, E. H. Postel, and D. M. Kaetzel. 2002. NM23-H1 and NM23-H2 repress transcriptional activities of nucleasehypersensitive elements in the platelet-derived growth factor-A promoter. J. Biol. Chem. 277:1560. 160. Jans, D. A., L. J. Briggs, P. Jans, C. J. Froelich, G. Parasivam, S. Kumar, V. R. Sutton, and J. A. Trapani. 1998. Nuclear targeting of the serine protease granzyme A (fragmentin-1). J. Cell Sci. 111 ( Pt 17):2645. 161. Jans, D. A., P. Jans, L. J. Briggs, V. Sutton, and J. A. Trapani. 1996. Nuclear transport of granzyme B (fragmentin-2). Dependence of perforin in vivo and cytosolic factors in vitro. J. Biol. Chem. 271:30781. 162. Zhang, D., M. S. Pasternack, P. J. Beresford, L. Wagner, A. H. Greenberg, and J. Lieberman. 2001. Induction of rapid histone degradation by the cytotoxic T lymphocyte protease Granzyme A. J. Biol. Chem. 276:3683. 163. Wilharm, E., J. Tschopp, and D. E. Jenne. 1999. Biological activities of granzyme K are conserved in the mouse and account for residual Z-Lys-SBzl activity in granzyme Adeficient mice. FEBS Lett. 459:139. 164. Babe, L. M., S. Yoast, M. Dreyer, and B. F. Schmidt. 1998. Heterologous expression of human granzyme K in Bacillus subtilis and characterization of its hydrolytic activity in vitro. Biotechnol. Appl. Biochem. 27 ( Pt 2):117. 54 165. Edwards, K. M., C. M. Kam, J. C. Powers, and J. A. Trapani. 1999. The human cytotoxic T cell granule serine protease granzyme H has chymotrypsin-like (chymase) activity and is taken up into cytoplasmic vesicles reminiscent of granzyme B-containing endosomes. J. Biol. Chem. 274:30468. 166. Sayers, T. J., A. D. Brooks, J. M. Ward, T. Hoshino, W. E. Bere, G. W. Wiegand, J. M. Kelly, M. J. Smyth, and J. M. Kelley. 2001. The restricted expression of granzyme M in human lymphocytes. J. Immunol. 166:765. 167. Mahrus, S., W. Kisiel, and C. S. Craik. 2004. Granzyme M is a regulatory protease that inactivates proteinase inhibitor 9, an endogenous inhibitor of granzyme B. J. Biol. Chem. 279:54275. 168. Sun, J., C. H. Bird, V. Sutton, L. McDonald, P. B. Coughlin, T. A. De Jong, J. A. Trapani, and P. I. Bird. 1996. A cytosolic granzyme B inhibitor related to the viral apoptotic regulator cytokine response modifier A is present in cytotoxic lymphocytes. J. Biol. Chem. 271:27802. 169. Medema, J. P., J. J. de, L. T. Peltenburg, E. M. Verdegaal, A. Gorter, S. A. Bres, K. L. Franken, M. Hahne, J. P. Albar, C. J. Melief, and R. Offringa. 2001. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc. Natl. Acad. Sci. U. S. A 98:11515. 170. Kelly, J. M., N. J. Waterhouse, E. Cretney, K. A. Browne, S. Ellis, J. A. Trapani, and M. J. Smyth. 2004. Granzyme M mediates a novel form of perforin-dependent cell death. J. Biol. Chem. 171. Andrin, C., M. J. Pinkoski, K. Burns, E. A. Atkinson, O. Krahenbuhl, D. Hudig, S. A. Fraser, U. Winkler, J. Tschopp, M. Opas, R. C. Bleackley, and M. Michalak. 1998. Interaction between a Ca2+-binding protein calreticulin and perforin, a component of the cytotoxic T-cell granules. Biochemistry 37:10386. 172. Fraser, S. A., M. Michalak, W. H. Welch, and D. Hudig. 1998. Calreticulin, a component of the endoplasmic reticulum and of cytotoxic lymphocyte granules, regulates perforinmediated lysis in the hemolytic model system. Biochem. Cell Biol. 76:881. 173. Woodard, S. L., S. A. Fraser, U. Winkler, D. S. Jackson, C. M. Kam, J. C. Powers, and D. Hudig. 1998. Purification and characterization of lymphocyte chymase I, a granzyme implicated in perforin-mediated lysis. J. Immunol. 160:4988. 174. Woodard, S. L., D. S. Jackson, A. S. Abuelyaman, J. C. Powers, U. Winkler, and D. Hudig. 1994. Chymase-directed serine protease inhibitor that reacts with a single 30-kDa granzyme and blocks NK-mediated cytotoxicity. J. Immunol. 153:5016. 175. Trapani, J. A., and M. J. Smyth. 2002. Functional significance of the perforin/granzyme cell death pathway. Nat. Rev. Immunol. 2:735. 55 176. Ebnet, K., M. Hausmann, F. Lehmann-Grube, A. Mullbacher, M. Kopf, M. Lamers, and M. M. Simon. 1995. Granzyme A-deficient mice retain potent cell-mediated cytotoxicity. EMBO J. 14:4230. 177. Heusel, J. W., R. L. Wesselschmidt, S. Shresta, J. H. Russell, and T. J. Ley. 1994. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76:977. 178. Mullbacher, A., P. Waring, H. R. Tha, T. Tran, S. Chin, T. Stehle, C. Museteanu, and M. M. Simon. 1999. Granzymes are the essential downstream effector molecules for the control of primary virus infections by cytolytic leukocytes. Proc. Natl. Acad. Sci. U. S. A 96:13950. 179. Revell, P. A., W. J. Grossman, D. A. Thomas, X. Cao, R. Behl, J. A. Ratner, Z. H. Lu, and T. J. Ley. 2005. Granzyme B and the Downstream Granzymes C and/or F Are Important for Cytotoxic Lymphocyte Functions. J. Immunol. 174:2124. 180. Riera, L., M. Gariglio, G. Valente, A. Mullbacher, C. Museteanu, S. Landolfo, and M. M. Simon. 2000. Murine cytomegalovirus replication in salivary glands is controlled by both perforin and granzymes during acute infection. Eur. J. Immunol. 30:1350. 181. Mullbacher, A., R. T. Hla, C. Museteanu, and M. M. Simon. 1999. Perforin is essential for control of ectromelia virus but not related poxviruses in mice. J. Virol. 73:1665. 182. Perona, J. J., and C. S. Craik. 1997. Evolutionary divergence of substrate specificity within the chymotrypsin-like serine protease fold. J. Biol. Chem. 272:29987. 183. Kam, C. M., D. Hudig, and J. C. Powers. 2000. Granzymes (lymphocyte serine proteases): characterization with natural and synthetic substrates and inhibitors. Biochim. Biophys. Acta 1477:307. 184. Caputo, A., M. N. James, J. C. Powers, D. Hudig, and R. C. Bleackley. 1994. Conversion of the substrate specificity of mouse proteinase granzyme B. Nat. Struct. Biol. 1:364. 185. Harris, J. L., E. P. Peterson, D. Hudig, N. A. Thornberry, and C. S. Craik. 1998. Definition and redesign of the extended substrate specificity of granzyme B. J. Biol. Chem. 273:27364. 186. Smyth, M. J., M. D. O' Connor, J. A. Trapani, M. H. Kershaw, and R. I. Brinkworth. 1996. A novel substrate-binding pocket interaction restricts the specificity of the human NK cell-specific serine protease, Met-ase-1. J. Immunol. 156:4174. 187. Powers, J. C., and C. M. Kam. 1995. Peptide thioester substrates for serine peptidases and metalloendopeptidases. Methods Enzymol. 248:3. 188. Farmer, D. A., and J. H. Hageman. 1975. Use of N-benzoyl-L-tyrosine thiobenzyl ester as a protease substrate. Hydrolysis by alpha-chymotrypsin and subtilisin BPN. J. Biol. Chem. 250:7366. 56 189. Hudig, D., N. J. Gregg, C. M. Kam, and J. C. Powers. 1987. Lymphocyte granulemediated cytolysis requires serine protease activity. Biochem. Biophys. Res. Commun. 149:882. 190. Beresford, P. J., C. M. Kam, J. C. Powers, and J. Lieberman. 1997. Recombinant human granzyme A binds to two putative HLA-associated proteins and cleaves one of them. Proc. Natl. Acad. Sci. U. S. A 94:9285. 191. Talanian, R. V., X. Yang, J. Turbov, P. Seth, T. Ghayur, C. A. Casiano, K. Orth, and C. J. Froelich. 1997. Granule-mediated killing: pathways for granzyme B-initiated apoptosis. J. Exp. Med. 186:1323. 192. Shi, L., C. M. Kam, J. C. Powers, R. Aebersold, and A. H. Greenberg. 1992. Purification of three cytotoxic lymphocyte granule serine proteases that induce apoptosis through distinct substrate and target cell interactions. J. Exp. Med. 176:1521. 193. Smyth, M. J., T. J. Sayers, T. Wiltrout, J. C. Powers, and J. A. Trapani. 1993. Met-ase: cloning and distinct chromosomal location of a serine protease preferentially expressed in human natural killer cells. J. Immunol. 151:6195. 194. Masson, D., and J. Tschopp. 1987. A family of serine esterases in lytic granules of cytolytic T lymphocytes. Cell 49:679. 195. Odake, S., C. M. Kam, L. Narasimhan, M. Poe, J. T. Blake, O. Krahenbuhl, J. Tschopp, and J. C. Powers. 1991. Human and murine cytotoxic T lymphocyte serine proteases: subsite mapping with peptide thioester substrates and inhibition of enzyme activity and cytolysis by isocoumarins. Biochemistry 30:2217. 196. Jackson, D. S., S. A. Fraser, L. M. Ni, C. M. Kam, U. Winkler, D. A. Johnson, C. J. Froelich, D. Hudig, and J. C. Powers. 1998. Synthesis and evaluation of diphenyl phosphonate esters as inhibitors of the trypsin-like granzymes A and K and mast cell tryptase. J. Med. Chem. 41:2289. 197. Poe, M., J. K. Wu, J. T. Blake, H. J. Zweerink, and N. H. Sigal. 1991. The enzymatic activity of human cytotoxic T-lymphocyte granzyme A and cytolysis mediated by cytotoxic T-lymphocytes are potently inhibited by a synthetic antiprotease, FUT-175. Arch. Biochem. Biophys. 284:215. 198. Andrade, F., H. G. Bull, N. A. Thornberry, G. W. Ketner, L. A. Casciola-Rosen, and A. Rosen. 2001. Adenovirus L4-100K assembly protein is a granzyme B substrate that potently inhibits granzyme B-mediated cell death. Immunity 14:751. 199. Trapani, J. A., and V. R. Sutton. 2003. Granzyme B: pro-apoptotic, antiviral and antitumor functions. Current Opinion in Immunology 15:533. 200. Johnson, H., L. Scorrano, S. J. Korsmeyer, and T. J. Ley. 2003. Cell death induced by granzyme C. Blood 101:3093. 57 201. Fan, Z., P. J. Beresford, D. Zhang, Z. Xu, C. D. Novina, A. Yoshida, Y. Pommier, and J. Lieberman. 2003. Cleaving the oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nat. Immunol. 4:145. 202. Szabo, C., and V. L. Dawson. 1998. Role of poly(ADP-ribose) synthetase in inflammation and ischaemia-reperfusion. Trends Pharmacol. Sci. 19:287. 203. Pinkoski, M. J., and D. R. Green. 2003. Granzyme A: the road less traveled. Nat. Immunol. 4:106. 204. Caputo, A., J. C. Parrish, M. N. James, J. C. Powers, and R. C. Bleackley. 1999. Electrostatic reversal of serine proteinase substrate specificity. Proteins 35:415. 205. Solivan, S., T. Selwood, Z. M. Wang, and N. M. Schechter. 2002. Evidence for diversity of substrate specificity among members of the chymase family of serine proteases. Febs Letters 512:133. 206. Bone, R., J. L. Silen, and D. A. Agard. 1989. Structural plasticity broadens the specificity of an engineered protease. Nature 339:191. 207. Tsu, C. A., J. J. Perona, R. J. Fletterick, and C. S. Craik. 1997. Structural basis for the broad substrate specificity of fiddler crab collagenolytic serine protease 1. Biochemistry 36:5393. 208. Wouters, M. A., K. Liu, P. Riek, and A. Husain. 2003. A despecialization step underlying evolution of a family of serine proteases. Mol. Cell 12:343. 209. Munford, R. S., P. O. Sheppard, and P. J. O' Hara. 1995. Saposin-like proteins (SAPLIP) carry out diverse functions on a common backbone structure. J. Lipid Res. 36:1653. 210. Pena, S. V., D. A. Hanson, B. A. Carr, T. J. Goralski, and A. M. Krensky. 1997. Processing, subcellular localization, and function of 519 (granulysin), a human late T cell activation molecule with homology to small, lytic, granule proteins. J. Immunol. 158:2680. 211. Jongstra, J., T. J. Schall, B. J. Dyer, C. Clayberger, J. Jorgensen, M. M. Davis, and A. M. Krensky. 1987. The isolation and sequence of a novel gene from a human functional T cell line. J. Exp. Med. 165:601. 212. Mincheva-Nilsson, L., O. Nagaeva, K. G. Sundqvist, M. L. Hammarstrom, S. Hammarstrom, and V. Baranov. 2000. gammadelta T cells of human early pregnancy decidua: evidence for cytotoxic potency. Int. Immunol. 12:585. 213. Gamen, S., D. A. Hanson, A. Kaspar, J. Naval, A. M. Krensky, and A. Anel. 1998. Granulysin-induced apoptosis. I. Involvement of at least two distinct pathways. J. Immunol. 161:1758. 58 214. Hanson, D. A., A. A. Kaspar, F. R. Poulain, and A. M. Krensky. 1999. Biosynthesis of granulysin, a novel cytolytic molecule. Mol. Immunol. 36:413. 215. Anderson, D. H., M. R. Sawaya, D. Cascio, W. Ernst, R. Modlin, A. Krensky, and D. Eisenberg. 2003. Granulysin crystal structure and a structure-derived lytic mechanism. J. Mol. Biol. 325:355. 216. Clayberger, C., and A. M. Krensky. 2003. Granulysin. Curr. Opin. Immunol. 15:560. 217. Hata, A., L. Zerboni, M. Sommer, A. A. Kaspar, C. Clayberger, A. M. Krensky, and A. M. Arvin. 2001. Granulysin blocks replication of varicella-zoster virus and triggers apoptosis of infected cells. Viral Immunol. 14:125. 218. Mackewicz, C. E., S. Ridha, and J. A. Levy. 2000. HIV virions and HIV replication are unaffected by granulysin. AIDS 14:328. 219. Pena, S. V., and A. M. Krensky. 1997. Granulysin, a new human cytolytic granuleassociated protein with possible involvement in cell-mediated cytotoxicity. Semin. Immunol. 9:117. 220. Kishi, A., Y. Takamori, K. Ogawa, S. Takano, S. Tomita, M. Tanigawa, M. Niman, T. Kishida, and S. Fujita. 2002. Differential expression of granulysin and perforin by NK cells in cancer patients and correlation of impaired granulysin expression with progression of cancer. Cancer Immunol. Immunother. 50:604. 221. Sekiya, M., A. Ohwada, M. Katae, T. Dambara, I. Nagaoka, and Y. Fukuchi. 2002. Adenovirus vector-mediated transfer of 9 kDa granulysin induces DNA fragmentation in HuD antigen-expressing small cell lung cancer murine model cells. Respirology. 7:29. 222. Kupfer, A., S. J. Singer, and G. Dennert. 1986. On the mechanism of unidirectional killing in mixtures of two cytotoxic T lymphocytes. Unidirectional polarization of cytoplasmic organelles and the membrane-associated cytoskeleton in the effector cell. J. Exp. Med. 163:489. 223. Balaji, K. N., N. Schaschke, W. Machleidt, M. Catalfamo, and P. A. Henkart. 2002. Surface cathepsin B protects cytotoxic lymphocytes from self-destruction after degranulation. Journal of Experimental Medicine 196:493. 224. Hirst, C. E., M. S. Buzza, C. H. Bird, H. S. Warren, P. U. Cameron, M. L. Zhang, P. G. shton-Rickardt, and P. I. Bird. 2003. The intracellular granzyme B inhibitor, proteinase inhibitor 9, is up-regulated during accessory cell maturation and effector cell degranulation, and its overexpression enhances CTL potency. Journal of Immunology 170:805. 225. Potempa, J., E. Korzus, and J. Travis. 1994. The serpin superfamily of proteinase inhibitors: structure, function, and regulation. J. Biol. Chem. 269:15957. 59 226. Poe, M., C. D. Bennett, W. E. Biddison, J. T. Blake, G. P. Norton, J. A. Rodkey, N. H. Sigal, R. V. Turner, J. K. Wu, and H. J. Zweerink. 1988. Human cytotoxic lymphocyte tryptase. Its purification from granules and the characterization of inhibitor and substrate specificity. J. Biol. Chem. 263:13215. 227. Masson, D., and J. Tschopp. 1988. Inhibition of lymphocyte protease granzyme A by antithrombin III. Mol. Immunol. 25:1283. 228. Bladergroen, B. A., M. C. Strik, N. Bovenschen, B. O. van, G. L. Scheffer, C. J. Meijer, C. E. Hack, and J. A. Kummer. 2001. The granzyme B inhibitor, protease inhibitor 9, is mainly expressed by dendritic cells and at immune-privileged sites. J. Immunol. 166:3218. 229. Bird, P. I. 1999. Regulation of pro-apoptotic leucocyte granule serine proteinases by intracellular serpins. Immunol. Cell Biol. 77:47. 230. Quan, L. T., A. Caputo, R. C. Bleackley, D. J. Pickup, and G. S. Salvesen. 1995. Granzyme B is inhibited by the cowpox virus serpin cytokine response modifier A. J. Biol. Chem. 270:10377. 231. Sun, J., L. Ooms, C. H. Bird, V. R. Sutton, J. A. Trapani, and P. I. Bird. 1997. A new family of 10 murine ovalbumin serpins includes two homologs of proteinase inhibitor 8 and two homologs of the granzyme B inhibitor (proteinase inhibitor 9). J. Biol. Chem. 272:15434. 232. Hill, R. M., K. S. Morresey, L. C. Coates, E. Mezey, B. Fell, T. Bratt, J. A. Trapani, and N. P. Birch. 1998. A new intracellular serine protease inhibitor expressed in the rat pituitary gland complexes with granzyme B. FEBS Lett. 440:361. 233. Granger, G. A., S. J. Shacks, T. W. Williams, and W. P. Kolb. 1969. Lymphocyte in vitro cytotoxicity: specific release of lymphotoxin-like materials from tuberculin-sensitive lymphoid cells. Nature 221:1155. 234. Carswell, E. A., L. J. Old, R. L. Kassel, S. Green, N. Fiore, and B. Williamson. 1975. An endotoxin-induced serum factor that causes necrosis of tumors. Proc. Natl. Acad. Sci. U. S. A 72:3666. 235. Baker, S. J., and E. P. Reddy. 1998. Modulation of life and death by the TNF receptor superfamily. Oncogene 17:3261. 236. Bodmer, J. L., P. Schneider, and J. Tschopp. 2002. The molecular architecture of the TNF superfamily. Trends Biochem. Sci. 27:19. 237. Locksley, R. M., N. Killeen, and M. J. Lenardo. 2001. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104:487. 238. Eck, M. J., and S. R. Sprang. 1989. The structure of tumor necrosis factor-alpha at 2.6 A resolution. Implications for receptor binding. J. Biol. Chem. 264:17595. 60 239. Jones, E. Y., D. I. Stuart, and N. P. Walker. 1989. Structure of tumour necrosis factor. Nature 338:225. 240. Banner, D. W., A. D' Arcy, W. Janes, R. Gentz, H. J. Schoenfeld, C. Broger, H. Loetscher, and W. Lesslauer. 1993. Crystal structure of the soluble human 55 kd TNF receptor-human TNF beta complex: implications for TNF receptor activation. Cell 73:431. 241. Beutler, B. 1990. TNF in pathophysiology: biosynthetic regulation. J. Invest Dermatol. 95:81S. 242. Kinkhabwala, M., P. Sehajpal, E. Skolnik, D. Smith, V. K. Sharma, H. Vlassara, A. Cerami, and M. Suthanthiran. 1990. A novel addition to the T cell repertory. Cell surface expression of tumor necrosis factor/cachectin by activated normal human T cells. J. Exp. Med. 171:941. 243. Monastra, G., A. Cabrelle, A. Zambon, A. Rosato, B. Macino, D. Collavo, and P. Zanovello. 1996. Membrane form of TNF alpha induces both cell lysis and apoptosis in susceptible target cells. Cell Immunol. 171:102. 244. Abe, Y., A. Horiuchi, Y. Osuka, S. Kimura, G. A. Granger, and T. Gatanaga. 1992. Studies of membrane-associated and soluble (secreted) lymphotoxin in human lymphokine-activated T-killer cells in vitro. Lymphokine Cytokine Res. 11:115. 245. Ware, C. F., P. D. Crowe, M. H. Grayson, M. J. Androlewicz, and J. L. Browning. 1992. Expression of surface lymphotoxin and tumor necrosis factor on activated T, B, and natural killer cells. J. Immunol. 149:3881. 246. Suda, T., T. Okazaki, Y. Naito, T. Yokota, N. Arai, S. Ozaki, K. Nakao, and S. Nagata. 1995. Expression of the Fas ligand in cells of T cell lineage. J. Immunol. 154:3806. 247. Lowin, B., M. Hahne, C. Mattmann, and J. Tschopp. 1994. Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways. Nature 370:650. 248. Kagi, D., F. Vignaux, B. Ledermann, K. Burki, V. Depraetere, S. Nagata, H. Hengartner, and P. Golstein. 1994. Fas and perforin pathways as major mechanisms of T cellmediated cytotoxicity. Science 265:528. 249. Berke, G. 1995. The CTL' s kiss of death. Cell 81:9. 250. Podack, E. R. 1995. Functional significance of two cytolytic pathways of cytotoxic T lymphocytes. J. Leukoc. Biol. 57:548. 251. Henkart, P. A. 1994. Lymphocyte-mediated cytotoxicity: two pathways and multiple effector molecules. Immunity 1:343. 252. Lopez-Cepero, M., J. A. Garcia-Sanz, L. Herbert, R. Riley, M. E. Handel, E. R. Podack, and D. M. Lopez. 1994. Soluble and membrane-bound TNF-alpha are involved in the 61 cytotoxic activity of B cells from tumor-bearing mice against tumor targets. J. Immunol. 152:3333. 253. Lee, R. K., J. Spielman, D. Y. Zhao, K. J. Olsen, and E. R. Podack. 1996. Perforin, Fas ligand, and tumor necrosis factor are the major cytotoxic molecules used by lymphokineactivated killer cells. J. Immunol. 157:1919. 254. Kashii, Y., R. Giorda, R. B. Herberman, T. L. Whiteside, and N. L. Vujanovic. 1999. Constitutive expression and role of the TNF family ligands in apoptotic killing of tumor cells by human NK cells. J. Immunol. 163:5358. 255. Vitolo, D., N. L. Vujanovic, H. Rabinowich, M. Schlesinger, R. B. Herberman, and T. L. Whiteside. 1993. Rapid Il-2-induced adherence of human natural killer cells. Expression of mRNA for cytokines and IL-2 receptors in adherent NK cells. J. Immunol. 151:1926. 256. Perez, C., I. Albert, K. DeFay, N. Zachariades, L. Gooding, and M. Kriegler. 1990. A nonsecretable cell surface mutant of tumor necrosis factor (TNF) kills by cell-to-cell contact. Cell 63:251. 257. Black, R. A., C. T. Rauch, C. J. Kozlosky, J. J. Peschon, J. L. Slack, M. F. Wolfson, B. J. Castner, K. L. Stocking, P. Reddy, S. Srinivasan, N. Nelson, N. Boiani, K. A. Schooley, M. Gerhart, R. Davis, J. N. Fitzner, R. S. Johnson, R. J. Paxton, C. J. March, and D. P. Cerretti. 1997. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 385:729. 258. Decker, T., M. L. Lohmann-Matthes, and G. E. Gifford. 1987. Cell-associated tumor necrosis factor (TNF) as a killing mechanism of activated cytotoxic macrophages. J. Immunol. 138:957. 259. Ratner, A., and W. R. Clark. 1993. Role of TNF-alpha in CD8+ cytotoxic T lymphocytemediated lysis. J. Immunol. 150:4303. 260. Jongeneel, C. V., S. A. Nedospasov, G. Plaetinck, P. Naquet, and J. C. Cerottini. 1988. Expression of the tumor necrosis factor locus is not necessary for the cytolytic activity of T lymphocytes. J. Immunol. 140:1916. 261. Sean, R. D., H. Korner, D. H. Strickland, F. A. Lemckert, J. D. Pollard, and J. D. Sedgwick. 1998. Challenging cytokine redundancy: inflammatory cell movement and clinical course of experimental autoimmune encephalomyelitis are normal in lymphotoxin-deficient, but not tumor necrosis factor-deficient, mice. J. Exp. Med. 187:1517. 262. Roths, J. B., E. D. Murphy, and E. M. Eicher. 1984. A new mutation, gld, that produces lymphoproliferation and autoimmunity in C3H/HeJ mice. J. Exp. Med. 159:1. 263. Cohen, P. L., and R. A. Eisenberg. 1991. Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease. Annu. Rev. Immunol. 9:243. 62 264. Smyth, M. J., and R. W. Johnstone. 2000. Role of TNF in lymphocyte-mediated cytotoxicity. Microsc. Res. Tech. 50:196. 265. Dick, T., G. Reichmann, K. Ebnet, M. M. Simon, H. P. Dienes, B. Echternacher, P. H. Krammer, and A. B. Reske-Kunz. 1993. An ovalbumin peptide-specific cytotoxic T cell clone with antigen self-presentation capacity uses two distinct mechanisms to kill target cells. Cell Immunol. 152:333. 266. Liu, A. N., A. Z. Mohammed, W. R. Rice, D. T. Fiedeldey, J. S. Liebermann, J. A. Whitsett, T. J. Braciale, and R. I. Enelow. 1999. Perforin-independent CD8(+) T-cellmediated cytotoxicity of alveolar epithelial cells is preferentially mediated by tumor necrosis factor-alpha: relative insensitivity to Fas ligand. Am. J. Respir. Cell Mol. Biol. 20:849. 267. Ando, K., K. Hiroishi, T. Kaneko, T. Moriyama, Y. Muto, N. Kayagaki, H. Yagita, K. Okumura, and M. Imawari. 1997. Perforin, Fas/Fas ligand, and TNF-alpha pathways as specific and bystander killing mechanisms of hepatitis C virus-specific human CTL. J. Immunol. 158:5283. 268. Gagnon, S. J., F. A. Ennis, and A. L. Rothman. 1999. Bystander target cell lysis and cytokine production by dengue virus-specific human CD4(+) cytotoxic T-lymphocyte clones. J. Virol. 73:3623. 269. Smyth, M. J., E. Krasovskis, and R. W. Johnstone. 1998. Fas ligand-mediated lysis of self bystander targets by human papillomavirus-specific CD8+ cytotoxic T lymphocytes. J. Virol. 72:5948. 270. Smyth, M. J., and J. D. Sedgwick. 1998. Delayed kinetics of tumor necrosis factormediated bystander lysis by peptide-specific CD8+ cytotoxic T lymphocytes. Eur. J. Immunol. 28:4162. 271. Qian, J. H., J. A. Titus, S. M. Andrew, D. Mezzanzanica, M. A. Garrido, J. R. Wunderlich, and D. M. Segal. 1991. Human peripheral blood lymphocytes targeted with bispecific antibodies release cytokines that are essential for inhibiting tumor growth. J. Immunol. 146:3250. 272. Rosendahl, A., K. Kristensson, J. Hansson, K. Riesbeck, T. Kalland, and M. Dohlsten. 1998. Perforin and IFN-gamma are involved in the antitumor effects of antibody-targeted superantigens. J. Immunol. 160:5309. 273. Nagata, S., and P. Golstein. 1995. The Fas death factor. Science 267:1449. 274. Combadiere, B., Reis e Sousa, C. Trageser, L. X. Zheng, C. R. Kim, and M. J. Lenardo. 1998. Differential TCR signaling regulates apoptosis and immunopathology during antigen responses in vivo. Immunity 9:305. 63 275. Martinez-Lorenzo, M. J., M. A. Alava, S. Gamen, K. J. Kim, A. Chuntharapai, A. Pineiro, J. Naval, and A. Anel. 1998. Involvement of APO2 ligand/TRAIL in activationinduced death of Jurkat and human peripheral blood T cells. Eur. J. Immunol. 28:2714. 276. Tucek-Szabo, C. L., S. Andjelic, E. Lacy, K. B. Elkon, and J. Nikolic-Zugic. 1996. Surface T cell Fas receptor/CD95 regulation, in vivo activation, and apoptosis. Activation-induced death can occur without Fas receptor. J. Immunol. 156:192. 277. Wang, J., S. A. Stohlman, and G. Dennert. 1994. TCR cross-linking induces CTL death via internal action of TNF. J. Immunol. 152:3824. 278. Zheng, L., G. Fisher, R. E. Miller, J. Peschon, D. H. Lynch, and M. J. Lenardo. 1995. Induction of apoptosis in mature T cells by tumour necrosis factor. Nature 377:348. 279. Speiser, D. E., E. Sebzda, T. Ohteki, M. F. Bachmann, K. Pfeffer, T. W. Mak, and P. S. Ohashi. 1996. Tumor necrosis factor receptor p55 mediates deletion of peripheral cytotoxic T lymphocytes in vivo. Eur. J. Immunol. 26:3055. 280. Singer, G. G., and A. K. Abbas. 1994. The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice. Immunity 1:365. 281. Sytwu, H. K., R. S. Liblau, and H. O. McDevitt. 1996. The roles of Fas/APO-1 (CD95) and TNF in antigen-induced programmed cell death in T cell receptor transgenic mice. Immunity 5:17. 282. Zhou, T., C. K. Edwards, III, P. Yang, Z. Wang, H. Bluethmann, and J. D. Mountz. 1996. Greatly accelerated lymphadenopathy and autoimmune disease in lpr mice lacking tumor necrosis factor receptor I. J. Immunol. 156:2661. 283. Deveraux, Q. L., and J. C. Reed. 1999. IAP family proteins--suppressors of apoptosis. Genes Dev. 13:239. 284. Rothe, M., M. G. Pan, W. J. Henzel, T. M. Ayres, and D. V. Goeddel. 1995. The TNFR2TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins. Cell 83:1243. 285. Uren, A. G., M. Pakusch, C. J. Hawkins, K. L. Puls, and D. L. Vaux. 1996. Cloning and expression of apoptosis inhibitory protein homologs that function to inhibit apoptosis and/or bind tumor necrosis factor receptor-associated factors. Proc. Natl. Acad. Sci. U. S. A 93:4974. 286. Haas, E., M. Grell, H. Wajant, and P. Scheurich. 1999. Continuous autotropic signaling by membrane-expressed tumor necrosis factor. J. Biol. Chem. 274:18107. 287. Schneider, P., N. Holler, J. L. Bodmer, M. Hahne, K. Frei, A. Fontana, and J. Tschopp. 1998. Conversion of membrane-bound Fas(CD95) ligand to its soluble form is associated with downregulation of its proapoptotic activity and loss of liver toxicity. J. Exp. Med. 187:1205. 64 288. Eissner, G., F. Kohlhuber, M. Grell, M. Ueffing, P. Scheurich, A. Hieke, G. Multhoff, G. W. Bornkamm, and E. Holler. 1995. Critical involvement of transmembrane tumor necrosis factor-alpha in endothelial programmed cell death mediated by ionizing radiation and bacterial endotoxin. Blood 86:4184. 289. Grell, M., E. Douni, H. Wajant, M. Lohden, M. Clauss, B. Maxeiner, S. Georgopoulos, W. Lesslauer, G. Kollias, K. Pfizenmaier, and . 1995. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83:793. 290. Tanaka, M., T. Itai, M. Adachi, and S. Nagata. 1998. Downregulation of Fas ligand by shedding. Nat. Med. 4:31. 291. Sarin, A., M. Conan-Cibotti, and P. A. Henkart. 1995. Cytotoxic effect of TNF and lymphotoxin on T lymphoblasts. J. Immunol. 155:3716. 292. Suda, T., H. Hashimoto, M. Tanaka, T. Ochi, and S. Nagata. 1997. Membrane Fas ligand kills human peripheral blood T lymphocytes, and soluble Fas ligand blocks the killing. J. Exp. Med. 186:2045. 293. Tartaglia, L. A., D. V. Goeddel, C. Reynolds, I. S. Figari, R. F. Weber, B. M. Fendly, and M. A. Palladino, Jr. 1993. Stimulation of human T-cell proliferation by specific activation of the 75-kDa tumor necrosis factor receptor. J. Immunol. 151:4637. 294. Lahn, M., H. Kalataradi, P. Mittelstadt, E. Pflum, M. Vollmer, C. Cady, A. Mukasa, A. T. Vella, D. Ikle, R. Harbeck, R. O' Brien, and W. Born. 1998. Early preferential stimulation of gamma delta T cells by TNF-alpha. J. Immunol. 160:5221. 295. Santis, A. G., M. R. Campanero, J. L. Alonso, and F. Sanchez-Madrid. 1992. Regulation of tumor necrosis factor (TNF)-alpha synthesis and TNF receptors expression in T lymphocytes through the CD2 activation pathway. Eur. J. Immunol. 22:3155. 296. Le, G. S., D. E. Le, N. Labarriere, J. F. Fonteneau, C. Viret, E. Diez, and F. Jotereau. 1998. LFA-3 co-stimulates cytokine secretion by cytotoxic T lymphocytes by providing a TCR-independent activation signal. Eur. J. Immunol. 28:1322. 297. Kuhweide, R., D. J. Van, and J. L. Ceuppens. 1990. Tumor necrosis factor-alpha and interleukin 6 synergistically induce T cell growth. Eur. J. Immunol. 20:1019. 298. Wallach, D., E. E. Varfolomeev, N. L. Malinin, Y. V. Goltsev, A. V. Kovalenko, and M. P. Boldin. 1999. Tumor necrosis factor receptor and Fas signaling mechanisms. Annu. Rev. Immunol. 17:331. 299. Baud, V., and M. Karin. 2001. Signal transduction by tumor necrosis factor and its relatives. Trends Cell Biol. 11:372. 65 300. Hsu, H., H. B. Shu, M. G. Pan, and D. V. Goeddel. 1996. TRADD-TRAF2 and TRADDFADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 84:299. 301. Rothe, M., S. C. Wong, W. J. Henzel, and D. V. Goeddel. 1994. A novel family of putative signal transducers associated with the cytoplasmic domain of the 75 kDa tumor necrosis factor receptor. Cell 78:681. 302. Rothe, M., V. Sarma, V. M. Dixit, and D. V. Goeddel. 1995. TRAF2-mediated activation of NF-kappa B by TNF receptor 2 and CD40. Science 269:1424. 303. Stanger, B. Z., P. Leder, T. H. Lee, E. Kim, and B. Seed. 1995. RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell 81:513. 304. Chinnaiyan, A. M., K. O' Rourke, M. Tewari, and V. M. Dixit. 1995. FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 81:505. 305. Duan, H., and V. M. Dixit. 1997. RAIDD is a new ' death'adaptor molecule. Nature 385:86. 306. Kelliher, M. A., S. Grimm, Y. Ishida, F. Kuo, B. Z. Stanger, and P. Leder. 1998. The death domain kinase RIP mediates the TNF-induced NF-kappaB signal. Immunity 8:297. 307. Nagata, S. 1997. Apoptosis by death factor. Cell 88:355. 308. Muzio, M., A. M. Chinnaiyan, F. C. Kischkel, K. O' Rourke, A. Shevchenko, J. Ni, C. Scaffidi, J. D. Bretz, M. Zhang, R. Gentz, M. Mann, P. H. Krammer, M. E. Peter, and V. M. Dixit. 1996. FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death--inducing signaling complex. Cell 85:817. 309. Boldin, M. P., T. M. Goncharov, Y. V. Goltsev, and D. Wallach. 1996. Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1- and TNF receptorinduced cell death. Cell 85:803. 310. Li, H., H. Zhu, C. J. Xu, and J. Yuan. 1998. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 94:491. 311. Zimmerman, R. J., A. Chan, and S. A. Leadon. 1989. Oxidative damage in murine tumor cells treated in vitro by recombinant human tumor necrosis factor. Cancer Res. 49:1644. 312. Grell, M., G. Zimmermann, E. Gottfried, C. M. Chen, U. Grunwald, D. C. Huang, Y. H. Wu Lee, H. Durkop, H. Engelmann, P. Scheurich, H. Wajant, and A. Strasser. 1999. Induction of cell death by tumour necrosis factor (TNF) receptor 2, CD40 and CD30: a role for TNF-R1 activation by endogenous membrane-anchored TNF. EMBO J. 18:3034. 66 313. Reinhard, C., B. Shamoon, V. Shyamala, and L. T. Williams. 1997. Tumor necrosis factor alpha-induced activation of c-jun N-terminal kinase is mediated by TRAF2. EMBO J. 16:1080. 314. Rathmell, J. C., and C. B. Thompson. 1999. The central effectors of cell death in the immune system. Annu. Rev. Immunol. 17:781. 315. Mixter, P. F., J. Q. Russell, G. J. Morrissette, C. Charland, D. eman-Hoey, and R. C. Budd. 1999. A model for the origin of TCR-alphabeta+ CD4-CD8- B220+ cells based on high affinity TCR signals. J. Immunol. 162:5747. 316. Cantrell, D. A., and K. A. Smith. 1984. The interleukin-2 T-cell system: a new cell growth model. Science 224:1312. 317. geciras-Schimnich, A., T. S. Griffith, D. H. Lynch, and C. V. Paya. 1999. Cell cycledependent regulation of FLIP levels and susceptibility to Fas-mediated apoptosis. J. Immunol. 162:5205. 318. Van, P. L., Y. Refaeli, J. D. Lord, B. H. Nelson, A. K. Abbas, and D. Baltimore. 1999. Uncoupling IL-2 signals that regulate T cell proliferation, survival, and Fas-mediated activation-induced cell death. Immunity 11:281. 319. Brunner, T., R. J. Mogil, D. LaFace, N. J. Yoo, A. Mahboubi, F. Echeverri, S. J. Martin, W. R. Force, D. H. Lynch, C. F. Ware, and . 1995. Cell-autonomous Fas (CD95)/Fasligand interaction mediates activation-induced apoptosis in T-cell hybridomas. Nature 373:441. 320. Bonfoco, E., P. M. Stuart, T. Brunner, T. Lin, T. S. Griffith, Y. Gao, H. Nakajima, P. A. Henkart, T. A. Ferguson, and D. R. Green. 1998. Inducible nonlymphoid expression of Fas ligand is responsible for superantigen-induced peripheral deletion of T cells. Immunity 9:711. 321. Hildeman, D. A., T. Mitchell, T. K. Teague, P. Henson, B. J. Day, J. Kappler, and P. C. Marrack. 1999. Reactive oxygen species regulate activation-induced T cell apoptosis. Immunity 10:735. 322. Ida, H., and P. Anderson. 1998. Activation-induced NK cell death triggered by CD2 stimulation. Eur. J. Immunol. 28:1292. 323. Ida, H., M. J. Robertson, S. Voss, J. Ritz, and P. Anderson. 1997. CD94 ligation induces apoptosis in a subset of IL-2-stimulated NK cells. J. Immunol. 159:2154. 324. Ida, H., T. Nakashima, N. L. Kedersha, S. Yamasaki, M. Huang, Y. Izumi, T. Miyashita, T. Origuchi, A. Kawakami, K. Migita, P. I. Bird, P. Anderson, and K. Eguchi. 2003. Granzyme B leakage-induced cell death: a new type of activation-induced natural killer cell death. Eur. J. Immunol. 33:3284. 67 325. Ullberg, M., and M. Jondal. 1981. Recycling and target binding capacity of human natural killer cells. J. Exp. Med. 153:615. 326. Chambers, C. A., T. J. Sullivan, and J. P. Allison. 1997. Lymphoproliferation in CTLA4-deficient mice is mediated by costimulation-dependent activation of CD4+ T cells. Immunity 7:885. 327. Boise, L. H., A. J. Minn, P. J. Noel, C. H. June, M. A. Accavitti, T. Lindsten, and C. B. Thompson. 1995. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity 3:87. 328. Kirchhoff, S., W. W. Muller, A. Krueger, I. Schmitz, and P. H. Krammer. 2000. TCRmediated up-regulation of c-FLIPshort correlates with resistance toward CD95-mediated apoptosis by blocking death-inducing signaling complex activity. J. Immunol. 165:6293. 329. Kirchhoff, S., W. W. Muller, M. Li-Weber, and P. H. Krammer. 2000. Up-regulation of c-FLIPshort and reduction of activation-induced cell death in CD28-costimulated human T cells. Eur. J. Immunol. 30:2765. 330. Salvesen, G. S., and C. S. Duckett. 2002. IAP proteins: blocking the road to death' s door. Nat. Rev. Mol. Cell Biol. 3:401. 331. Johnson, D. E., B. R. Gastman, E. Wieckowski, G. Q. Wang, A. Amoscato, S. M. Delach, and H. Rabinowich. 2000. Inhibitor of apoptosis protein hILP undergoes caspasemediated cleavage during T lymphocyte apoptosis. Cancer Res. 60:1818. 332. Deveraux, Q. L., N. Roy, H. R. Stennicke, A. T. Van, Q. Zhou, S. M. Srinivasula, E. S. Alnemri, G. S. Salvesen, and J. C. Reed. 1998. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J. 17:2215. 333. Roy, N., Q. L. Deveraux, R. Takahashi, G. S. Salvesen, and J. C. Reed. 1997. The c-IAP1 and c-IAP-2 proteins are direct inhibitors of specific caspases. EMBO J. 16:6914. 334. Deveraux, Q. L., R. Takahashi, G. S. Salvesen, and J. C. Reed. 1997. X-linked IAP is a direct inhibitor of cell-death proteases. Nature 388:300. 335. Yang, X., H. Y. Chang, and D. Baltimore. 1998. Autoproteolytic activation of procaspases by oligomerization. Mol. Cell 1:319. 336. Clem, R. J., T. T. Sheu, B. W. Richter, W. W. He, N. A. Thornberry, C. S. Duckett, and J. M. Hardwick. 2001. c-IAP1 is cleaved by caspases to produce a proapoptotic Cterminal fragment. J. Biol. Chem. 276:7602. 337. Wang, C. Y., M. W. Mayo, R. G. Korneluk, D. V. Goeddel, and A. S. Baldwin, Jr. 1998. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281:1680. 68 338. Partheniou, F., S. M. Kelsey, S. M. Srinivasula, A. C. Newland, E. S. Alnemri, and L. Jia. 2001. c-IAP1 blocks TNFalpha-mediated cytotoxicity upstream of caspase-dependent and -independent mitochondrial events in human leukemic cells. Biochem. Biophys. Res. Commun. 287:181. 339. Joazeiro, C. A., and A. M. Weissman. 2000. RING finger proteins: mediators of ubiquitin ligase activity. Cell 102:549. 340. Huang, H., C. A. Joazeiro, E. Bonfoco, S. Kamada, J. D. Leverson, and T. Hunter. 2000. The inhibitor of apoptosis, cIAP2, functions as a ubiquitin-protein ligase and promotes in vitro monoubiquitination of caspases 3 and 7. J. Biol. Chem. 275:26661. 341. Suzuki, Y., Y. Imai, H. Nakayama, K. Takahashi, K. Takio, and R. Takahashi. 2001. A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol. Cell 8:613. 342. LeBlanc, A. C. 2003. Natural cellular inhibitors of caspases. Prog. Neuropsychopharmacol. Biol. Psychiatry 27:215. 343. Brinkmann, U., E. Brinkmann, M. Gallo, and I. Pastan. 1995. Cloning and Characterization of A Cellular Apoptosis Susceptibility Gene, the Human Homolog to the Yeast Chromosome Segregation Gene Cse1. Proceedings of the National Academy of Sciences of the United States of America 92:10427. 344. Brinkmann, U., E. Brinkmann, M. Gallo, U. Scherf, and I. Pastan. 1996. Role of CAS, a human homologue to the yeast chromosome segregation gene CSE1, in toxin and tumor necrosis factor mediated apoptosis. Biochemistry 35:6891. 345. Brinkmann, U., M. Gallo, M. H. Polymeropoulos, and I. Pastan. 1996. The human CAS (cellular apoptosis susceptibility) gene mapping on chromosome 20q13 is amplified in BT474 breast cancer cells and part of aberrant chromosomes in breast and colon cancer cell lines. Genome Research 6:187. 346. Brinkmann, U. 1998. CAS, the human homologue of the yeast chromosome-segregation gene CSE1, in proliferation, apoptosis, and cancer. Am. J. Hum. Genet. 62:509. 347. Kutay, U., F. R. Bischoff, S. Kostka, R. Kraft, and D. Gorlich. 1997. Export of importin alpha from the nucleus is mediated by a specific nuclear transport factor. Cell 90:1061. 348. Scherf, U., P. Kalab, M. Dasso, I. Pastan, and U. Brinkmann. 1998. The hCSE1/CAS protein is phosphorylated by HeLa extracts and MEK-1: MEK-1 phosphorylation may modulate the intracellular localization of CAS. Biochem. Biophys. Res. Commun. 250:623. 349. Behrens, P., U. Brinkmann, F. Fogt, N. Wernert, and A. Wellmann. 2001. Implication of the proliferation and apoptosis associated CSE1L/CAS gene for breast cancer development. Anticancer Res. 21:2413. 69 350. Takenaka, K., Y. Gotoh, and E. Nishida. 1997. MAP kinase is required for the spindle assembly checkpoint but is dispensable for the normal M phase entry and exit in Xenopus egg cell cycle extracts. J. Cell Biol. 136:1091. 351. Stilo, R., D. Liguoro, J. B. di, A. Leonardi, and P. Vito. 2003. The alpha-chain of the nascent polypeptide-associated complex binds to and regulates FADD function. Biochem. Biophys. Res. Commun. 303:1034. 352. Al-Shanti, N., C. G. Steward, R. J. Garland, and A. W. Rowbottom. 2004. Investigation of alpha nascent polypeptide-associated complex functions in a human CD8(+) T cell ex vivo expansion model using antisense oligonucleotides. Immunology 112:397. 353. Inohara, N., and G. Nunez. 2000. Genes with homology to mammalian apoptosis regulators identified in zebrafish. Cell Death. Differ. 7:509. 354. Evans, D. L., S. L. Taylor, J. H. Leary, III, G. R. Bishop, A. Eldar, and L. JasoFriedmann. 2000. In vivo activation of tilapia nonspecific cytotoxic cells by Streptococcus iniae and amplification with apoptosis regulatory factor(s). Fish. Shellfish. Immunol. 10:419. 355. Evans, D. L., J. H. Leary, III, and L. Jaso-Friedmann. 2001. Nonspecific cytotoxic cells and innate immunity: regulation by programmed cell death. Dev. Comp Immunol. 25:791. 356. Evans, D. L., R. L. Carlson, S. S. Graves, and K. T. Hogan. 1984. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). IV. Target cell binding and recycling capacity. Dev. Comp Immunol. 8:823. 70 CHAPTER 3 EVIDENCE FOR THE EXISTENCE OF GRANZYME-LIKE SERINE PROTEASES IN TELEOST CYTOTOXIC CELLS1 1 Praveen, K., D. L. Evans and L. Jaso-Friedmann. 2004. Journal of Molecular Evolution. 58:449-459. © Springer-Verlag New York, LLC 2004. Reprinted here with permission of publisher. The original publication is available at http://www.springerlink.com/app/home/contribution.asp?wasp=n2gwppwryr3bf3wn9h2m&refer rer=parent&backto=issue,8,11;journal,9,109;linkingpublicationresults,1:100107,1 71 ABSTRACT Granzymes are granule associated serine proteases, which are important effector molecules in NK cell and CTL functions. The granzyme family poses a perplexing problem in phylogenetics due to the lack of non-mammalian sequence information. We now report the identification of a cDNA that codes for a granzyme homologue, channel catfish granzyme-1 (CFGR-1) from nonspecific cytotoxic cells (NCC) of a teleost. NCC are the first identified and extensively studied cytotoxic cell population in teleosts. Ictalurus punctatus (channel catfish) granzyme cDNA encodes a protein with ~50% similarity to granzymes A and K. Highly conserved catalytic triad residues of serine proteases and other motifs common to granzymes were also identified. Conserved amino acid sequences, structure-function data available for serine protease family and the crystal structure of human granzyme K supported a model of CFGR-1. It suggested an Arg/Lys primary substrate specificity that is shared with granzyme A and K. Furthermore, CFGR-1 has the four conserved disulfide bonds of granzyme A, K, and M. Phylogenetic analysis suggested that this molecule is a member of the granzyme family. Expression of CFGR-1 in NCC was confirmed by RT-PCR analysis. Presence of a granzymelike molecule that might play an important role in the effector functions of NCC indicates that cell-mediated immunity with granule exocytosis and Fas pathways have been conserved for more than 300 millions of years. Key words: NK cells, Cytotoxic T cells, Granzymes, Cytotoxicity, Granule exocytosis, Comparative immunology/ Evolution. 72 INTRODUCTION Mammalian cytotoxic T cells (CTL) and natural killer (NK) cells play an important role in immunosurveillance against virus-infected and tumor cells. Although recognition of target cells differs greatly, the mechanisms by which CTL and NK cells cause target cell death following formation of conjugates appear to be the same. Two main cytotoxic pathways have been described that are independent from each other (Waterhouse and Trapani 2002). One pathway involves the crosslinking of death receptors on the target cell (members of the TNF receptor family such as Fas) by death ligands present on effector cells (FasL, TRAIL). This interaction leads to apoptotic target cell death (Trenn et al. 1987; Kagi et al. 1994). The other pathway requires granule exocytosis with the resulting secretion of two abundant granular proteins, perforin and granzymes, which appear to act in concert to induce target cell death (Henkart 1985; Podack 1985; Young and Cohn 1986; Peters et al. 1990; Podack and Kupfer 1991; Doherty 1993). Both pathways act synergistically on many target cells, but the exocytosis pathway alone is sufficient to kill target cells, which do not express death receptors. The mechanisms of action of perforin and granzymes have been the subject of intense studies (Henkart 1985; Podack 1985; Young and Cohn 1986; Peters et al. 1990; Podack and Kupfer 1991; Doherty 1993). Perforin is a pore-forming molecule that in addition to inducing the lysis of cells by osmotic leakage acts synergistically with granzymes to facilitate their entry into target cells. Granzymes are members of a granule-associated serine proteases family expressed in CTL and NK cells that induce apoptosis of target cells either through caspase activation or through caspase-independent pathways. Recent evidence suggests that even in the absence of perforin, granzymes can enter target cells by receptor-mediated endocytosis via the cationindependent mannose 6-phosphate receptor. However, internalization of granzyme in the absence 73 of perforin is without toxic consequences (Froelich et al. 1996; Jans et al. 1998; Browne et al. 1999; Motyka et al. 2000). Four orthologous members of the granzyme family with different substrate specificities have been identified in humans and rodents (A, B, K, M) and additional non-orthologous genes exist in the human (H), rat (J) and mouse (C, D, E, F, G, N) genome (Trapani 2001; Barry and Bleackley 2002). The granzyme loci have been placed into three groups by their chromosomal location and structural similarities. They have four primary specificities including "aspase", "chymase", "tryptase" and "metase" referring to the type of activities of the proteases encoded for in their genes. Granzymes A and B have different substrate specificities (tryptase and aspase, respectively) but they are both pro-apoptotic. Although the function of Granzyme M, a metase, has not been determined, it is believed to play a role in NK cell function as it is only expressed in those cytotoxic cells. The substrate specificity of other family members remains unknown. The diversity of enzymatic activities of the granzyme family members has prompted speculation that these proteases may play a role in other immune functions such as lymphocyte migration and extravasation as well as amplification of cytokine function (Simon et al. 1991; Sayers et al. 1992; Vettel et al. 1993; Smyth et al. 2001). It is apparent that in spite of their biological importance, many questions remain unanswered about granzymes. At present, only murine, rat and human sequence information for granzymes is available and the identification of evolutionary precursors would provide an important step in the elucidation of their biological activities. This is the first report of the characterization of a non-mammalian granzyme sequenced from lower vertebrates. Nonspecific cytotoxic cells (NCC) are the teleost equivalent to mammalian NK cells. Since their initial description in the channel catfish (Graves et al. 1985; Evans et al. 1988; Jaso- 74 Friedmann et al. 1990; Evans et al. 1998; Evans and Jaso-Friedmann 1999), these cells have been well characterized in a number of lower vertebrates (Faisal et al. 1989; Greenlee et al. 1991; McKinney and Schmale 1994; Suzumura et al. 1994; Jaso-Friedmann and Evans 1999; JasoFriedmann et al. 2002). Although originally described as agranular lymphocytes (Graves et al. 1985), NCC from catfish, tilapia and trout have subsequently been shown to kill target cells by granule exocytosis and use both necrotic and apoptotic pathways (Faisal et al. 1989; JasoFriedmann et al. 1990; Greenlee et al. 1991). We have previously reported the importance of the Fas ligand-mediated killing mechanism of mammalian target cells by NCC (Bishop et al. 2000; Jaso-Friedmann et al. 2000). While others have also reported that catfish cytotoxic T cell lines kill target cells by the granzyme-perforin pathway, the lack of molecular tools did not allow the authors to identify these proteins (Zhou et al. 2001). In the present study we have identified a granzyme from a cDNA library constructed from cytotoxic cells of the lower vertebrate Ictalurus punctatus. Sequence analysis showed that this novel teleost protein shares highest similarity with the human and murine granzymes K and A. Primers designed from the sequenced enzyme in catfish were used to identify another granzyme homologue from NCC of tilapia (Oreocromus niloticus). Based on the phylogenetic sequence analysis we conclude that granzyme-like homologs already exist in teleost fishes. Characterization of granzyme precursors from lower vertebrates in turn will advance our understanding of the multiple roles of granzymes in the mammalian immune system. 75 MATERIALS AND METHODS Experimental animals and isolation of NCC. Outbred channel catfish (Ictalurus punctatus) of both sexes weighing 10-25 g were obtained from local commercial farms and outbred tilapia (Oreochromis niloticus) of both sexes weighing 60-100 g were obtained from Americulture, Inc. Animas, NM. Fish were maintained in fiberglass aquaria equipped with a constant flow through system at ambient water temperatures for channel catfish and 23-25o C for tilapia. Tilapia and catfish were fed a commercial diet of pelleted fish food (Southern States Co-operative Inc, VA). All animals were acclimatized for a minimum of 3 months prior to experimentation and were free from any active infections. NCC were purified from anterior kidney (bone marrow equivalent in fish) of channel catfish and peripheral blood of tilapia as described before (Jaso-Friedmann et al. 1990; Bishop et al. 2000). Purity of cell preparation was verified by flow cytometric analysis using 5C6 (a monoclonal antibody detecting NCCRP-1, which is an activation marker found exclusively on NCC, Evans et al. 1998, Jaso-Friedmann et al. 2002). Construction of cDNA library Total RNA was isolated from purified NCC with TRIzol® (Invitrogen) and mRNA was purified by the PolyATtract® mRNA isolation system (Promega). The purified mRNA was used to construct an expression cDNA library using ZAP-cDNA® Gigapack® III Gold Cloning kit (Stratagene) according to manufacture's instructions. The library was amplified and recombinant phages were converted into pBluescript plamids by in vivo excision according to manufacture's instructions. This was used as template for the PCR reactions in the initial screening for granzyme cDNA. 76 Screening for granzyme cDNA The strategy used in sequencing granzyme mRNA of channel catfish is illustrated in Figure 1. Multiple sequence alignments were done using Clustal W provided with vector NTI package, version 6 (InforMax Inc) to identify conserved domains in the known mammalian granzymes. Primers were designed (Table 1) and polymerase chain reactions were done to amplify specific regions of the cDNA. YT-INDY, a human natural killer-like leukemic cell line (Montel et al. 1995) cDNA was used as a positive control. Amplicons were TA cloned in to pDrive cloning vector using a PCR cloning kit (Qiagen). Inserts were sequenced in two directions and compared with the known sequences in DDBJ/EMBL/GenBank databases using BLAST version 2.2.5 (Altschul et al. 1997). Rapid amplification of cDNA ends Sequence from one of the amplicon which had closest similarity to the known granzyme sequences was used to design primers to amplify the entire 5' and 3' ends of the catfish granzyme cDNA. Fresh RNA was purified from catfish anterior kidney NCC for the RACE. For 5' RACE, purified mRNA (500 ng) was reverse transcribed using granzyme specific primers using Generacer Superscript II RT module (Invitrogen). First strand cDNA was purified using PCR purification kit (Qiagen) followed by homopolymeric tailing with Cytosine using terminal transferace (Roche). Tailed cDNA was purified using PCR purification kit (Qiagen). Later dCtailed cDNA was amplified using abridged anchor primer and gene specific nested primer followed by a re-amplification using a nested abridged universal amplification primer (AUAP) and another nested gene-specific primer. The amplicons were purified by gel extraction kit (Qiagen) and TA cloned for sequencing. For 3' RACE, catfish anterior kidney RNA was reverse 77 transcribed using an anchor primer, EPB-18T (Evans et al. 1998) to generate first strand cDNA. Using gene specific nested primers and EPB the 3' end of the mRNA was amplified and TA cloned for sequencing. The sequences were edited and assembled to obtain the complete sequence of catfish granzyme. Later forward and reverse primers were designed at the two ends of the open reading frame to amplify the coding region, which was TA cloned to verify the sequences. Phylogenetic analysis Similar analyses as those previously done with NCCRP-1 of zebrafish were performed (Jaso-Friedmann et al. 2002). Briefly, multiple sequence alignments were done using Clustal W, using only the mature portion of various proteases. Phylogenetic analysis was done using Mega version 2.1 (Kumar et al. 2001). The aligned data set was analyzed by the criteria of maximum parsimony using the branch-and-bound algorithm. The reliability of the trees was tested using 1000 bootstrap replicates. The alignment was also analyzed by the neighbor joining method as implemented by Mega with 1000 bootstrap replications. For neighbor joining method, Poisson correction was used with the complete deletion option. The tree was rooted on a sub tree consisting of trypsin and chymotrypsin sequences to resolve the differences between highly similar granzyme sequences. Protein modeling The three dimensional structure of catfish granzyme was modeled using SWISS-MODEL in the first approach mode accessible via the internet (http://www.expasy.org/swissmod). The known crystal structures of serine proteases whose crystal structure has been resolved and had 78 high sequence identity upon pairwise alignments (protein databank entries 1MZA (Hink-Schauer et al. 2002): human granzyme K, 1DST (Jing et al. 1998): complement factor D) were used as template for the modeling. The co-ordinate files were imported to RasWin software version 2.6 for analyzing bond lengths and other conformational features of the molecule. RT-PCR Total RNA was extracted from channel catfish NCC (from anterior kidney, spleen and peripheral blood), unfractionated head kidney, spleen, blood, gill, heart, muscle, and trunk kidney using TRIizol® (Life Technologies). The purified RNA (3 µg) was reverse transcribed in to cDNA using Generacer Superscript II RT module (Invitrogen). The final volume of cDNA synthesis reaction was 50 µl and 2 µl of the same was used in a 50 µl PCR reaction. Beta-actin was used as a normalization control for RT-PCR. PCR was done with initial denaturation at 94o C for 2 minutes followed by 30 cycles as follows: 30 s denaturation at 94o C, 30 s annealing at 57o C, and 30 s extension at 72o C. The products were resolved on a 1.5% agarose gel and visualized by ethidium bromide staining. RESULTS Nucleotide and deduced amino acid sequence of catfish granzyme An expression cDNA library generated from catfish anterior kidney NCC was used as template with different primers designed based on conserved (functional) domains in human and murine granzymes. YT-INDY cDNA was used in the PCR reactions as a positive control. The amplicons were analyzed on 1.5% agarose gel. Among the different products, one with identical size to a product generated by the same primer set from YT cDNA was selected for sequencing. 79 The sequence was compared and verified for the similarity to known granzymes. Based on the obtained product, additional non - degenerate primers were synthesized to obtain the sequence for the complete transcript by 5'- and 3'-RACE. The transcript of the catfish granzyme cDNA was 978 base pairs (bp) in length, with a single open reading frame yielding 768 bp (Fig. 2). One putative start codon was identified near the 5' end of the full-length cDNA determined by 5' RACE. The catfish granzyme cDNA encodes a putative mature protein with 231 amino acids. The predicated molecular mass of the unglycosylated protein was calculated as 25,526 daltons, with an isoelectric point of 9.56, suggesting highly basic nature of the enzyme. Initial analysis of the translated cDNA sequence by comparison to other known granzymes in the database suggested that the product was a member of the granzyme family of serine proteases (Figure 2). The catfish sequence was subsequently submitted to GenBank (Accession number: AY286012). The three key amino acid residues representing the catalytic triad (charge relay system) of serine proteases as well as their neighboring residues are well conserved in catfish granzyme (Figure 2, boxes). The three residues, His at position 57, Asp at 102 and Ser at 195 (chymotrypsinogen numbering system) are in similar locations as those of other serine proteases. A summary of these results showed that the mature granzyme sequence from catfish had 29-43% identity and 39-52% similarity to other granzymes (Table 2). Primary sequence analysis for identification of signature motifs of known granzymes A signal sequence of 20 amino acids was identified at the N-terminal end indicating that this enzyme is sorted to the secretory pathway (Figure 3). The mature protein begins at +1 with a highly conserved Ile-Ile-Gly-Gly motif found in the majority of the granzymes. The most likely cleavage site for the signal sequence was predicted between residues at -5 (Cys) and -4 (Ser) as 80 determined by weight matrix analysis of von Heijne (von Heijne 1986). There is a single putative glycosylation site at the N-terminus of the mature protein (Asn20-Asn-Ser). Prediction of tertiary structure and substrate specificity The three dimensional structure of catfish granzyme was modeled using SWIS-MODEL. The templates used for modeling were based on known crystal structures of serine proteases. Disulfide linkages are crucial in correct folding of serine proteases and their catalytic ability. The mature catfish granzyme has an unusually high number of Cys residues (11 in total). Out of these, 6 are highly conserved in all serine proteases (positions 42, 58, 136, 168, 182 and 201: chymotrypsinogen numbering). By analogy and based on results from other granzymes, the formation of disulfide bonds can be predicted as follows: 42 with 58; 136 with 168; 182 with 201). Cys191 and Cys220 (chymotrypsinogen numbering) could be analogous to a fourth disulfide linkage which bridges active site serine, like in chymotrypsin. Additional comparisons with other granzymes yielded more similarities like the formation of an internal salt bridge between the characteristic N-terminal motif (IIGG) and Aspchymotrypsinogen # 194 . The overall structure of the predicted model for catfish granzyme was similar to other granzymes, especially granzyme K (data not shown). The model was also instrumental to predict the active site conformation of catfish granzyme. Based on the structure of trypsin (Rypniewski et al. 1994), the residues which determine the configuration of active site were identified in the model (Gly43, Gly44, Gly140, Trp141, Gly142 and Leu155; chymotrypsinogen numbering). Those residues are in close proximity of the catalytic triad suggesting a highly conserved trypsin like active site structure. Four different enzymatic activities (tryptase, chymase, aspase and metase) have been assigned to the granzymes identified so far. The combination of amino acid residues that are 81 found to determine the primary substrate specificity of granzymes (-6, +16, +17 and +18 relative to active site Serine) were analyzed in catfish granzyme by examining the primary sequence and predicted model. The most critical residues determining the S1 subsite are found at positions 189, 216 and 226 (chymotrypsin numbering). Comparing these residues to that e.g. in granzyme A, B, cathepsin G and leukocyte elastase clearly indicates a Lys/Arg specificity (like granzyme A and K) for CFGR-1 (Jackson et al. 1998). Phylogenetic analysis Initial multiple alignments and visual examination of the catfish granzyme exposed many conserved domains common to the mammalian granzymes. The aligned data set was analyzed by different methods to obtain reliable phylogenetic data. Similar results were obtained by the methods used and a representative tree is shown in Figure 4. The similarity to multiple granzymes at different regions of the molecule posed problems for the grouping with the known enzymes. In order to resolve the differences between highly similar granzyme sequences, the tree was rooted on a sub-tree consisting of trypsin and chymotrypsin sequences. Catfish granzyme did not cluster with any of the granzyme families, but branched out as a separate group, indicating that the three major granzyme branches (granzyme AK, granzyme BCDEFGH, granzyme M/elastase-2/proteinase 3/factor D) started to diverge just before or after the emergence of teleost fishes. Expression pattern of the transcript for the catfish granzyme The presence of granzyme transcripts were analyzed in different tissue. RT-PCR was performed with primers designed based on the least conserved sequences between the teleost 82 granzymes and other known granzymes in order to selectively obtain the predicted amplicon. Figure 5A shows a representative experiment suggesting that tissue with highest concentration of NCC (head kidney, spleen and liver) had a higher level of granzyme mRNA than whole blood, muscle and heart tissue. However, there was significant increase in the expression levels of granzymes when the NCC were enriched from the whole blood (Figure 5B), indicating that NCC are the main source of CFGR-1 in catfish. Expression of granzymes in other fish species An expression cDNA library was constructed from tilapia NCC purified from peripheral blood. An initial primer pair, which amplified a 330 bp amplicon from catfish library, was used to screen the tilapia library by PCR. An amplicon of similar size as obtained from catfish and YT-INDY cDNA was sequenced. The sequence obtained was compared with known sequences in DDBJ/EMBL/GenBank databases using BLAST version 2.2.5. The PCR product had high similarity to known granzyme sequences. The tilapia granzyme PCR product was translated and aligned with catfish granzyme using Clustal W (Figure 6, panel A). There are considerable differences between the corresponding regions of the two molecules (36% identity and 46% similarity between the corresponding regions), but at the same time tilapia sequence retains the signature motifs common to all granzymes. This is suggestive evidence for the presence of different members of the granzyme family in other fish species. In order to identify potential representatives of granzyme family in other fish species, the catfish cDNA was compared with sequences in the EST database of NCBI. A Salmo salar EST (Accession number CB516537) was found to be similar to the catfish granzyme sequence. The EST sequence was edited and translated to obtain the predicted protein sequence. Although this EST is lacking a few residues 83 at both the ends and also the two untranslated regions, considerable similarity (53% identity and 63% similarity) between the catfish granzyme molecule and the translation of the EST was found (Figure 6, panel B). Additional searches showed that NCBI database has listed a partial sequence of a granzyme-like molecule from Salmo salar (accession number AF434669), similar to human granzyme M. The N-terminal portion of the mature protein from that sequence was compared with catfish granzyme. The N-terminal 43 amino acid domain of that protein has 56% identity and 65% similarity to the corresponding region of catfish granzyme (Figure 6, panel C). DISCUSSION Although several laboratories have functionally shown the existence of the granzyme/perforin pathway in the cytotoxic activity of teleost CTL and NK-like cells (Greenlee et al. 1991; Evans and Jaso-Friedmann 1999; Zhou et al. 2001), this is the first report of the identification of a non-mammalian granzyme in catfish and tilapia NCC. The primary sequence characteristics that place this protein in the granzyme family include the presence of signature motifs shown in Figures 2 and 3: the catalytic triad amino acids each of them with surrounding conserved residues; the presence of a propeptide that must be removed to convert the inactive zymogen in to an active protease, the phylogenetically conserved N-terminus amino acids; the cysteine residues important in disulphide bond formation; and the necessary leader sequence to place the nascent protein into the endoplasmic reticulum. The novel enzyme's predicted tertiary structure is consistent with serine proteases and phylogenetic analysis indicates that CFGR-1 may be a member of granzyme family (Figure 4). Furthermore, we show that the expression of the catfish granzyme is restricted to lymphoid tissue (Figures 5). 84 Homology comparisons with known mammalian granzymes alone were not enough to determine the substrate specificity of catfish granzyme because of the lack of predominant similarities to a particular member of the family. However, based on residue identities and residue similarities (table 2), CFGR-1 appears to be most similar to the mammalian granzymes A and K. Both of these proteases are the only granzymes with tryptase activity sequenced to date. Support for the idea that the teleost granzyme cleavage sites may be after arginine and lysine was suggested by analyzing the putative substrate specificity pocket amino acids, which were determined by comparison to similar residues in other granzymes. The catalytic triad residues (histidine, aspartic acid and serine) are conserved across different granzyme species, suggesting similar active site structure. VLTAAHC around His 57 and GDSGGPL around Ser 195 are highly conserved, while DIML is conserved in many granzymes. Interestingly, the tilapia product had closer similarity to granzymes from the metase family. Although, it is still early to conclusively assign substrate specificity to this tilapia granzyme, the significance of this finding is that there may be a fish ortholog to granzyme M. Granzyme M is uniquely expressed by NK cells, which suggests that the NCC are the NK-like cells (or their precursors) in teleost. Other indication that these two teleost granzymes may belong to different families are that RT-PCR using catfish granzyme specific primers failed to amplify the tilapia cDNA. In addition to the charge relay system, other granzyme signature motifs are present in the catfish homologue. As has been shown in all mammalian granzymes (Kam et al. 2000; Smyth et al. 1996), the catfish enzyme is produced as a propeptide. The leader sequence is a necessary requirement for the translated message of these enzymes to be compartmentalized to the rough endoplasmic reticulum and their final destination in the exocytic granules (Jenne and Tschopp 1988; Bleackley et al. 1988). Application of von Heijne's algorithm would leave a tetrapeptide as 85 the inactivating portion of the enzyme. Although the propetide portion of most granzymes is a dipeptide, the only mammalian granzyme that is exclusively expressed in NK cells, granzyme M, has a hexapeptide at this position (Smyth et al. 1993). Mammalian granzymes vary significantly in the degree of glycosylation from none (granzyme C) to accounting as much as 50% of their total mass (granzyme D). The possibility of glycosylation for the catfish granzyme is interesting in that it could have functional significance. It has been shown that granzyme A and B are glycosylated with phosphorylated mannose rich residues, which recognize the mannose-6-phosphate receptor. This is an important pathway for targeting of these enzymes to secretory granules. Moreover, glycosylation could allow target cell entry of the catfish granzymes in the absence of perforin by binding to the mannose-6-P receptor followed by endocytosis, an important point since the presence of teleost perforin has not been reported. Although the actual glycosylation status of the catfish granzyme is not presently known, it is noteworthy that the predicted protein has at least one putative glycosylation site at position 36 (chymotrypsinogen numbering). It has been demonstrated that glycosylation of granzymes occurs preferentially at Asn residues rather than Ser-Thr residues (Griffiths and Isaaz 1993). Based on the sequence information and expression pattern, this novel molecule can be predicted as a new member of granzyme family. This is the first report of a non-mammalian granzyme and there is convincing evidence for the presence of more granzyme-like molecules in fish cytotoxic cells. By definition, granzymes are found within the granules of cytotoxic cells. Characterization of granzymes from lower vertebrates sheds more light in to the evolutionary patterns of serine proteases. 86 ACKNOWLEDGEMENTS We thank Dr. David Peterson for critical review of the manuscript. This work was funded with support from USDA (98-35205-6701). REFERENCES Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ (1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res 25:3389-3402 Barry M, Bleackley RC (2002) Cytotoxic lymphocytes: All roads lead to death. Nat Rev Immunol 2:401- 409 Bishop GR, Jaso-Friedmann L, Evans DL (2000) Activation induced programmed cell death of nonspecific cytotoxic cells and inhibition by apoptosis regulatory factors. Cell Immunol 199:126-137 Bleackley RC, Lobe CG, Duggan B, Ehrman N, Fregeau C, Meier M, Letellier M, Havele C, Shaw J, Paetkau V (1988) The isolation and characterization of a family of serine protease genes expressed in activated cytotoxic T lymphocytes. Immunol Rev 103:5-19 Browne KA, Blink E, Sutton VR, Froelich CJ, Jans DA, Trapani JA (1999) Cytosolic delivery of granzyme B by bacterial toxins: evidence that endosomal disruption in addition to transmembrane pore formation is an important function of perforin. Mol Cell Biol 19:8604-8615 Doherty PC (1993) Cell-mediated cytotoxicity. Cell 75:607-612 Evans DL, Jaso-Friedmann L (1999) Nonspecific cytotoxic cells and innate cellular immunity in teleost fish. In Fingerman M , Nagabhushanam R (eds) Recent Advances in Marine Biotechnology Vol 5. Science Publishers, Inc. Enfield, pp. 243-268 Evans DL, Jaso-Friedmann L, Smith EE, St John A, Koren HS, Harris DT (1988) Identification of a putative antigen receptor on fish nonspecific cytotoxic cells with monoclonal antibodies. J Immunol 141:324-332 Evans DL, Leary JH, Jaso-Friedmann L (1998) NCCRP-1: a novel type III membrane protein on the teleost equivalent of natural killer cells recognizes conventional antigen. Cell Immunol 187:19-26 Faisal M, Ahmed II, Peters G, Cooper EL (1989) Natural cytotoxicity of tilapia leucocytes. Dis Aquat Organ 7:17-22 87 Froelich CJ, Orth K, Turbov J, Seth P, Gottlieb R, Babior B, Shah GM, Bleackley RC, Dixit VM, Hanna W (1996) New paradigm for lymphocyte granule-mediated cytotoxicity. Target cells bind and internalize granzyme B, but an endosomolytic agent is necessary for cytosolic delivery and subsequent apoptosis. J Biol Chem 271:29073-29079 Graves SS, Evans DL, Dawe DL (l985) Antiprotozoan activity of nonspecific cytotoxic cells (NCC) from the channel catfish (Ictalurus punctatus). J Immunol l34:78-85 Greenlee AR, Brown RA, Ristow SS (1991) Nonspecific cytotoxic cells of rainbow trout (Oncorhynchus mykiss) kill YAC-1 targets by both necrotic and apoptotic mechanisms. Dev Comp Immunol 15:153-164 Griffiths GM, Isaaz S (1993) Granzymes A and B are targeted to the lytic granules of lymphocytes by the mannose-6-phosphate receptor. J Cell Biol 120:885-896 Henkart PA (1985) Mechanism of lymphocyte-mediated cytotoxicity. Annu Rev Immunol 3:3158 Hink-Schauer C, Estebanez-Perpina E, Wilharm E, Fuentes-Prior P, Klinkert W, Bode W, Jenne DE (2002) The 2.2-A crystal structure of human pro-granzyme K reveals a rigid zymogen with unusual features. J Biol Chem 277:50923-50933 Jackson DS, Fraser SA, Ni LM, Kam CM, Winkler U, Johnson DA, Froelich CJ, Hudig D, Powers JC (1998) Synthesis and evaluation of diphenyl phosphonate esters as inhibitors of the trypsin-like granzymes A and K and mast cell tryptase. J Med Chem 41:2289-2301 Jans DA, Briggs LJ, Jans P, Froelich CJ, Parasivam G, Kumar S, Sutton VR, Trapani JA (1998) Nuclear targeting of the serine protease granzyme A (fragmentin-1). J Cell Sci 111:26452654 Jaso-Friedmann L, Evans DL (1999) Mechanisms of cellular cytotoxic innate resistance in tilapia (Oreochromis nilotica). Dev Comp Immunol. 23:27-35 Jaso-Friedmann L, Harris DT, St John A, Koren HS, Evans DL (1990) A monoclonal antibodypurified soluble target cell antigen inhibits nonspecific cytotoxic cell activity. J Immunol 144:2413-2418 Jaso-Friedmann L, Leary JH, Evans DL (2000) Role of nonspecific cytotoxic cells in the induction of programmed cell death of pathogenic protozoans: participation of the Fas ligand-Fas receptor system. Exp Parasitol 96:75-88 Jaso-Friedmann L, Peterson DS, Gonzalez DS, Evans DL (2002) The antigen receptor (NCCRP1) on catfish and zebrafish nonspecific cytotoxic cells belongs to a new gene family characterized by an F-box-associated domain. J Mol Evol 54:386-395 88 Jenne DE, Tschopp J (1988) Granzymes, a family of serine proteases released from granules of cytolytic T lymphocytes upon T cell receptor stimulation. Immunol Rev 103:53-71 Jing H, Babu YS, Moore D, Kilpatrick JM, Liu XY, Volanakis JE, Narayana SV (1998) Structures of native and complexed complement factor D: implications of the atypical His57 conformation and self-inhibitory loop in the regulation of specific serine protease activity. J Mol Biol 282:1061-1081 Kagi D, Ledermann B, Burki K, Seiler P, Odermatt B, Olsen KJ, Podack ER, Zinkernagel RM, Hengartner H (1994) Cytotoxicity mediated by T cells and natural killer cells is greatly impaired in perforin-deficient mice. Nature 369:31-37 Kam CM, Hudig D, Powers JC (2000) Lymphocyte serine proteases: characterization with natural and synthetic substrates and inhibitors. Biochim Biophys Acta 1477:307-323 Kumar S, Tamura K, Jakobsen IB, Nei M (2001) MEGA2: molecular evolutionary genetics analysis software. Bioinformatics 17:1244-1245 McKinney EC, Schmale MC (1994) Damselfish with neurofibromatosis exhibit cytotoxicity toward tumor targets. Dev Comp Immunol 18:305-313 Montel AH, Morse PA, Brahmi Z (1995) Upregulation of B7 molecules by the Epstein-Barr virus enhances susceptibility to lysis by a human NK-like cell line. Cell Immunol 160:104-114 Motyka B, Korbutt G, Pinkoski MJ, Heibein JA, Caputo A, Hobman M, Barry M, Shostak I, Sawchuk T, Holmes CF, Gauldie J, Bleackley RC (2000) Mannose-6 phosphate/insulinlike growth factor if receptor is a death receptor for granzyme B during cytotoxic T-cell induced apoptosis. Cell 103:491-500 Peters PJ, Geuze HJ, van der Donk HA, Borst J (1990) A new model for lethal hit delivery by cytotoxic T lymphocytes. Immunol Today 11:28-32 Podack ER (1985) The molecular mechanism of lymphocyte-mediated tumor cell lysis. Immunol Today 1:21-27 Podack ER, Kupfer A (1991) T-cell effector functions: mechanisms for delivery of cytotoxicity and help. Annu Rev Cell Biol 7:479-504 Rypniewski WR, Perrakis A, Vorgias CE, Wilson KS (1994) Evolutionary divergence and conservation of trypsin. Protein Eng 7:57-64 Sayers TJ, Wiltrout TA, Sowder R, Munger WL, Smyth MJ, Henderson LE (1992) Purification of a factor from the granules of a rate natural killer cell line (RNK) that reduces tumor cell growth and changes tumor morphology. Molecular identity with a granule serine protease (RNKP-1). J Immunol 148:292-300 89 Simon MM, Kramer MD, Prester M, Gay S (1991) Mouse T-cell associated serine protease I degrades collagen type IV: a structural basis for the migration of lymphocytes through vascular basement membrane. Immunology 73:117-119 Smyth MJ, Kelly JM, Sutton VR, Davis JE, Browne KA, Sayers TJ, Trapani JA (2001) Unlocking the secrets of cytotoxic granule proteins. J Leukoc Biol 70:18-29 Smyth MJ, O'Connor MD, Trapani JA (1996) Granzymes: a variety of serine protease specificities by genetically distinct subfamilies. J Leuk Biol 60: 555-562 Smyth MJ, Sayers TJ, Wiltrout T, Powers JC, Trapani JA (1993) Met-ase: cloning and distinct chromosomal location of a serine protease preferentially expressed in human natural killer cells. J Immunol 151:6195-6205 Suzumura E, Kurata O, Okamoto N, Ikeda Y (1994) Characteristics of natural killer-like cells in carp. Fish Path 29:199-203 Trapani JA (2001) Granzymes: a family of lymphocyte granule serine proteases. Genome Biol 2:3014.1. Trenn G, Takayama H, Sitkovsky MV (1987) Exocytosis of cytolytic granules may not be required for target cell lysis by cytotoxic T-lymphocytes. Nature 330:72-74 Vettel U, Brunner G, Bar-Shavit R, Vlodavsky I, Kramer MD (1993) Charge-dependent binding of granzyme A (MTSP-1) to basement membranes. Eur J Immunol 23:279-282. von Heijne G (1986) A new method for predicting signal sequence cleavage sites. Nucleic Acids Res 14:4683-4690 Waterhouse NJ, Trapani JA (2002) CTL: Caspases Terminate Life, but that's not the whole story. Tissue Antigens 59:179-183 Young JD, Cohn ZA (1986) Cell-mediated killing: a common mechanism? Cell 46:641-642 Zhou H, Stuge TB, Miller NW, Bengten E, Naftel JP, Bernanke J, Chinchar VG, Clem LW, Wilson M (2001) Heterogeneity of channel catfish CTL with respect to target recognition and cytotoxic mechanisms employed. J Immunol 167:1325-1332 90 Figure 3.1. Cloning strategy used to identify and characterize the cDNA for catfish granzyme. The primer sequences are listed in table 1. 91 92 Figure 3.2. Compiled full-length catfish granzyme cDNA sequence. Predicted signal sequence at the N-terminus is underlined. Putative tetrapeptide propeptide is double underlined. The amino acid numbering starts at the amino terminal of predicted mature protein as determined by sequence homology to other granzymes. Highly conserved IIXG motif at the N-terminal is boxed, while a region similar to the PHSRPYMA motif in other granzymes is highlighted. Putative glycosylation site is circled. Cysteine residues in the mature protein are marked with (•), while catalytic triad residues are marked with (♦). ATTTA motif in the 3' UTR is italicized with underline and poly A tail is underlined. 93 M 1 E R N T L K V K Q A T A S C G S -2 E K V K P K P W M A S 16 S H I C G G T L I H Q 32 A A H C K T F L Q F K P 49 L G A H S L T K D K N A M 66 V L C F H I S P K F S A T 82 H D I M L L K L Q D K V Q L 99 N K V D V K K I P K S G K D I 116 A G T K C E V R G W G T T H V 132 CCCAGCAGGGACAAAATGTGAAGTAAGAGGGTGGGGAACCACTCATGTAA P K A C D T L Q E L E V T V V 149 AAAATCCTAAGGCGTGTGACACCTTGCAAGAGCTGGAGGTGACGGTGGTG R E L C N C Y Y N S K P T I T A 166 GACAGGGAACTGTGTAACTGCTACTACAACAGCAAACCTACCATCACTGC M L C A G N K Q R D K D A C W 182 CAACATGCTGTGTGCAGGAAACAAGCAGAGGGACAAAGATGCATGCTGGG D S G G P L E C K K N I V G V V 199 GGGATTCTGGCGGACCTCTGGAGTGTAAGAAAAACATTGTGGGTGTGGTG S 701 F AAGAAAAACAAGGTAGACGTTAAGAAGATTCCCAAATCAGGCAAAGACAT G 651 L CTAGGGTTCATGACATCATGCTCCTGAAGCTCCAAGACAAAGTCCAGCTT N 601 T GCGAGTGAAAGTCCTATGCTTTCACATCTCACCAAAGTTCAGTGCTACAA D 551 G N L V V K N 501 L ATCGAAGTGCTTCTCGGTGCTCACTCTCTGACGAAGGACAAAAATGCTAT P 451 -19 AGTGGGTTCTGACAGCTGCACACTGCAAAACTTTTTTACAATTTAAACCA K 401 I G S V T R 351 L I Q W R 301 R AGTCCAGTCCAATAACTCGCACATCTGTGGAGGAACTCTTATTCATCAGC I 251 Q TTCATCATAGGAGGCCGAGAGGTGAAAAAACCTAAACCATGGATGGCCTC Q 201 L I V 151 Q CCCTTCTTCTGATTCTCACCTTGTTTCAAGCAACAGCTTGCTCAGGCAGT F 101 V CAGTCCTCATTTCTGCACTCAAGAGCAGACCATGCACGTGCAACAACGTT S L 51 H G G S G C G N P K K P G V Y T L TCAGGAGGAAGTGGCTGTGGCAATCCCAAAAAACCTGGTGTCTATACTCT L S K E H I D W I N K I I K K * 751 TCTCTCAAAGGAACACATTGACTGGATCAACAAGATAATTAAAAAGTAAT 801 CCAACAGCACAGAGGCTTAGAAACACAACATTTATCAGTGCTTGCAGTAT 851 ATATTGTGTCATATACAGTATCTGTCCGCATATACAAGAGGTATATTTGC 901 TTGCTTGTTGGTTTTATCTGATCTTTTACTTCACACATATAGCTGAAATT 951 GAACAGTACAGCAAAAAAAAAAAAAAAAn 94 216 Figure 3.3. Multiple sequence alignment of predicted catfish granzyme sequence (mature protein) with known granzymes and related proteins using CLUSTAL W. Highly conserved regions around the catalytic triad residues are indicated as boxes above the alignment. Cysteine residues marked (♦) are highly conserved among all known granzymes, while those marked (•) are conserved among granzymes A, K and M. Residues marked with (*) are predicted to contribute towards the primary substrate specificity of the protease. Mature portion of the following proteases were used in this comparison: human granzyme K (accession No. AAH35802), mouse granzyme K (accession No. AAC17930), rat granzyme K (accession No. NP_058815), human granzyme A (accession No. AAH15739), mouse granzyme A (accession No. NP_034500), rat granzyme M (accession No. Q03238), human granzyme M (accession No. AAH25701), mouse granzyme M (accession No. NP_032530), human granzyme B (accession No. AAH30195), mouse granzyme B (accession No. NP_038570), rat granzyme B (accession No. NP_612526), human granzyme H (accession No. A32692), mouse granzyme C (accession No. NP_034501), mouse granzyme D (accession No. NP_034502), mouse granzyme E (accession No. NP_034503), mouse granzyme F (accession No. AAA37741), mouse granzyme G (accession No. NP_034505), rat granzyme J (accession No. AAC53168), human mast cell chymase (accession No. KYHUCM), and human cathepsin G (NP_001902). Darker shading represents identical or residues with similar properties in 100% of the sequences and lighter shading represents identical or residues with similar properties in 80% or more of all the sequences. 95 CFGR-1 hum grnzK mus grnzK rat grnzK hum grnzA mus grnzA rat grnzM hum grnzM mus grnzM hum grnzB mus grnzB rat grnzB hum grnzH mus grnzC mus grnzD mus grnzE mus grnzF mus grnzG rat grnzJ hu mast chymase hu cathepsinG : : : : : : : : : : : : : : : : : : : : : IIGGREVK-KPKPWMASVQSNN----SHICGGTLIHQQWVLTAAHCKTFLQ-FKPIEVLL IIGGKEVSPHSRPFMASIQYGG----HHVCGGVLIDPQWVLTAAHCQYRFTKGQSPTVVL IIGGREVQPHSRPFMASIQYRS----KHICGGVLIHPQWVLTAAHCYSWFPRGHSPTVVL IIGGREVQPHSRPFMASIQYRG----KHICGGVLIHPQWVLTAAHCYS---RGHSPTVVL IIGGNEVTPHSRPYMVLLSLDR----KTICAGALIAKDWVLTAAHCNLN----KRSQVIL IIGGDTVVPHSRPYMALLKLSS----NTICAGALIEKNWVLTAAHCNVG----KRSKFIL IIGGREAVPHSRPYMVSLQNTK----SHMCGGVLVHQKWVLTAAHCLSEP--LQQLKLVF IIGGREVIPHSRPYMASLQRNG----SHLCGGVLVHPKWVLTAAHCLAQR--MAQLRLVL IIGGREAVPHSRPYMASLQKAK----SHVCGGVLVHRKWVLTAAHCLSEP--LQNLKLVL IIGGHEAKPHSRPYMAYLMIWDQKS-LKRCGGFLIQDDFVLTAAHCWGS-----SINVTL IIGGHEVKPHSRPYMALLSIKDQQP-EAICGGFLIREDFVLTAAHCEGS-----IINVTL IIGGHEADPHSRPYMAYLQYKNEDSRDTICGGFLIREDFVLTAAHCSGS-----KINVTL IIGGHEAKPHSRPYMAFVQFLQEKS-RKRCGGILVRKDFVLTAAHCQGS-----SINVTL IIGGNEISPHSRPYMAYYEFLKVGGKKMFCGGFLVRDKFVLTAAHCKGR-----SMTVTL IIGGHVVKPHSRPYMAFVMSVDIKGNRIYCGGFLIQDDFVLTAAHCKNS-----SMTVTL IIGGHVVKPHSRPYMAFVKSVDIEGNRRYCGGFLVQDDFVLTAAHCRNR-----TMTVTL IIGGHEVKPHSRPYMARVRFVKDNGKRHSCGGFLVQDYFVLTAAHCTGS-----SMRVIL IIGGHEVKPHSRPYMAFIKSVDIEGKKKYCGGFLVQDDFVLTAAHCRNR-----SMTVTL IIWGTESKPHSRPYMAFINFYDSNSDLNRCGGFLVAKDIVMTAAHCNGR-----NIKVIL IIGGTECKPHSRPYMAYLEIVTSNGPSKFCGGFLIRRNFVLTAAHCAGR-----SITVTL IIGGRESRPHSRPYMAYLQIQSPAGQS-RCGGFLVREDFVLTAAHCWGS-----NINVTL : : : : : : : : : : : : : : : : : : : : : 54 56 56 53 52 52 54 54 54 54 54 55 54 55 55 55 55 55 55 55 54 CFGR-1 hum grnzK mus grnzK rat grnzK hum grnzA mus grnzA rat grnzM hum grnzM mus grnzM hum grnzB mus grnzB rat grnzB hum grnzH mus grnzC mus grnzD mus grnzE mus grnzF mus grnzG rat grnzJ hu mast chymase hu cathepsinG : : : : : : : : : : : : : : : : : : : : : GAHSLTKDKNAMRVKVLCFHISPKFSAT--TRVHDIMLLKLQDKVQLKKNKVDVKKIPKS GAHSLSKNEASKQTLEIKKFIPFSRVTS-DPQSNDIMLVKLQTAAKLNKH-VKMLHIRSK GAHSLSKNEPMKQTFEIKKFIPFSRLQS-GSASHDIMLIKLRTAAELNKN-VQLLHLGSK GAHSLSKNEPMKQTFEIKEFIPFSGFKS-G--TNDIMLIKLRTAAELNKH-VQLLHLRSK GAHSITREEPTKQIMLVKKEFPYPCYDP-ATREGDLKLLQLTEKAKINKY-VTILHLPKK GAHSINKE-PEQQILTVKKAFPYPCYDE-YTREGDLQLVRLKKKATVNRN-VAILHLPKK GLHSLHDPQDPGLTFYIKQAIKHPGYN--LKYENDLALLKLDGRVKPSKN-VKPLALPRK GLHTLDS---PGLTFHIKAAIQHPRYKPVPALENDLALLQLDGKVKPSRT-IRPLALPSK GLHNLHDLQDPGLTFYIREAIKHPGYN--HKYENDLALLKLDRRVQPSKN-VKPLALPRK GAHNIKEQEPTQQFIPVKRPIPHPAYNP-KNFSNDIMLLQLERKAKRTRA-VQPLRLPSN GAHNIKEQEKTQQVIPMVKCIPHPDYNP-KTFSNDIMLLKLKSKAKRTRA-VRPLNLPRR GAHNIKEQEKTQQVIPVVKIIPHPAYNA-KTISNDIMLLKLKSKAKRTRA-VKTLSLPRS GAHNIKEQERTQQFIPVKRPIPHPAYNP-KNFSNDIMLLQLERKAKWTTA-VRPLRLPSS GAHNIKAKEETQQIIPVAKAIPHPDYNP-DDRSNDIMLLKLVRNAKRTRA-VRPLNLPRR GAHNITAKEETQQIIPVAKDIPHPDYNA-TIFYSDIMLLKLESKAKRTKA-VRPLKLPRS GAHNIKAKEETQQIIPVAKAIPHPDYNA-TAFFSDIMLLKLESKAKRTKA-VRPLKLPRP GAHNIRAKEETQQIIPVAKAIPHPAYDD-KDNTSDIMLLKLESKAKRTKA-VRPLKLPRP GAHNIKAKEETQQIIPVAKAIPHPAFNR-KHGTNDIMLLKLESKAKRTKA-VRPLKLPRP GAHNIKKRE-NTQVISVLKAKPHENFNS-DSLVNDIMLLKLERKAQLNGV-VKTIALPRS GAHNITEEEDTWQKLEVIKQFRHPKYNT-STLHHDIMLLKLKEKASLTLA-VGTLPFPSQ GAHNIQRRENTQQHITARRAIRHPQYNQ-RTIQNDIMLLQLSRRVRRNRN-VNPVALPRA : : : : : : : : : : : : : : : : : : : : : 112 114 114 109 110 109 111 110 111 112 112 113 112 113 113 113 113 113 112 113 112 CFGR-1 hum grnzK mus grnzK rat grnzK hum grnzA mus grnzA rat grnzM hum grnzM mus grnzM hum grnzB mus grnzB rat grnzB hum grnzH mus grnzC mus grnzD mus grnzE mus grnzF mus grnzG rat grnzJ hu mast chymase hu cathepsinG : : : : : : : : : : : : : : : : : : : : : -GKDIPAGTKCEVRGWGTTHVKNPKACDTLQELEVTVVDRELCNCYYN--SKPTITANML --TSLRSGTKCKVTGWGATDPDSLRPSDTLREVTVTVLSRKLCNSQSYYNGDPFITKDMV --NYLRDGTKCQVTGWGTTKPDLLTASDTLREVTVTIISRKRCNSQSYYNHKPVITKDMI --NYIRDGTKCQVTGWGSTKPDVLTTSDTLQEVTVTIISRKRCNSQSYYNHKPVITKDMI -GDDVKPGTMCQVAGWGRTH-NSASWSDTLREVNITIIDRKVCNDRNHYNFNPVIGMNMV -GDDVKPGTRCRVAGWGRFG-NKSAPSETLREVNITVIDRKICNDEKHYNFHPVIGLNMI PRDKPAEGSRCSTAGWGITHQ-RGQLAKSLQELDLRLLDTRMCNNSRFWNGVLTDS--ML -RQVVAAGTRCSMAGWGLTHQ-GGRLSRVLRELDLQVLDTRMCNNSRFWNGSLSPS--MV PRSKPAEGTWCSTAGWGMTHQ-GGPRARALQELDLRVLDTQMCNNSRFWNGVLIDS--ML -KAQVKPGQTCSVAGWGQTAP-LGKHSHTLQEVKMTVQEDRKCESDLRHYYDSTI---EL -NVNVKPGDVCYVAGWGRMAP-MGKYSNTLQEVELTVQKDRECESYFKNRYNKTN---QI -NFKVKPGDVCYVAGWGKLGP-MGKFPDKLQEVELTVQEDQECETYLKNAYDKAN---QI -KAQVKPGQLCSVAGWGYVS--MSTLATTLQEVLLTVQKDCQCERLFHGNYSRAT---EI -NAHVKPGDECYVAGWGKVTP-DGEFPKTLHEVKLTVQKDQVCESQFQSSYNRAN---EI -NARVKPGDVCSVAGWGSRSINDTKASARLREVQLVIQEDEECKKRFR-YYTETT---EI -NARVKPGDVCSVAGWGPRSINDTKASARLREAQLVIQEDEECKKRFR-HYTETT---EI -NARVKPGHVCSVAGWGRTSINATQRSSCLREAQLIIQKDKECKKYFY-KYFKTM---QI -NARVKPGDVCSVAGWGKTSINATKASARLREAQLIIQEDEECKKLWY-TYSKTT---QI -QDWVKPGQVCTVAGWGTLA--NCTLSNTLQEVNLEVQKGQKCQGMSR-NYNDSI---QL -FNFVPPGRMCRVAGWGRTG-VLKPGSDTLQEVKLRLMDPQACSHFRD--FDHNL---QL -QEGLRPGTLCTVAGWGRVS--MRRGTDTLREVQLRVQRDRQCLRIFG-SYDPRR---QI : : : : : : : : : : : : : : : : : : : : : 169 172 172 167 168 167 168 166 168 167 167 168 166 168 168 168 168 168 165 166 165 96 * *** CFGR-1 hum grnzK mus grnzK rat grnzK hum grnzA mus grnzA rat grnzM hum grnzM mus grnzM hum grnzB mus grnzB rat grnzB hum grnzH mus grnzC mus grnzD mus grnzE mus grnzF mus grnzG rat grnzJ hu mast chymase hu cathepsinG : : : : : : : : : : : : : : : : : : : : : CAGNKQRDKDACWGDSGGPLECKKN--IVGVVSGGSG--CGNPKKPGVYTLLSKEHIDWI CAGDAKGQKDSCKGDSGGPLICKGV--FHAIVSGGHE--CGVATKPGIYTLLTKKYQTWI CAGDARGQKDSCKGDSGGPLICKGI--FHALVSQGYK--CGIAKKPGIYTLLTKKYQTWI CAGDRRGEKDSCKGDSGGPLICKGV--FHALVSGGYK--CGISNKPGVYTLLTKKYQTWI CAGSLRGGRDSCNGDSGSPLLCEGV--FRGVTSFGLENKCGDPRGPGVYILLSKKHLNWI CAGDLRGGKDSCNGDSGSPLLCDGI--LRGITSFGGE-KCGDRRWPGVYTFLSDKHLNWI CLKAGAKGQAPCKGDSGGPLVCGKG-KVDGILSFSSK-NCTD-IFKPTVATAVAPYSSWI CLAADSKDQAPCKGDSGGPLVCGKGRVLAGVLSFSSR-VCTD-IFKPPVATAVAPYVSWI CLKAGSKSQAPCKGDSGGPLVCGKG-QVDGILSFSSK-TCTD-IFKPPVATAVAPYSSWI CVGDPEIKKTSFKGDSGGPLVCNKV--AQGIVSYGRN-----NGMPPRACTKVSSFVHWI CAGDPKTKRASFRGDSGGPLVCKKV--AAGIVSYGYK-----DGSPPRAFTKVSSFLSWI CAGDPKIKCASFQGDSGGPLVCKKV--AAGIVSYGRK-----DGSTPRAFTKVSTFLSWI CVGDPKKTQTGFKGDSGGPLVCKDV--AQGILSYGNK-----KGTPPGVYIKVSHFLPWI CVGDSKIKGASFEEDSGGPLVCKRA--AAGIVSYGQT-----DGSAPQVFTRVLSFVSWI CAGDLKKIKTPFKGDSGGPLVCHNQ--AYGLFAYAKN-----GTISSGIFTKVVHFLPWI CAGDLKKIKTPFKGDSGGPLVCDNK--AYGLLAYAKN-----RTISSGVFTKIVHFLPWI CAGDPKKIQSTYSGDSGGPLVCNNK--AYGVLTYGLN-----RTIGPGVFTKVVHYLPWI CAGDPKKVQAPYEGESGGPLVCDNL--AYGVVSYGIN-----RTITPGVFTKVVHFLPWI CVGNPNERKATAGGDSGGPFVCNGV--AQGIVSYRLC-----TWTPPRVFTRISSFIPWI CVGNPRKTKSAFKGDSGGPLLCAGV--AQGIVSYGRS-----DAKPPAVFTRISHYRPWI CVGDRRERKAAFKGDSGGPLLCNNV--AHGIVSYGKS-----SGVPPEVFTRVSSFLPWI CFGR-1 hum grnzK mus grnzK rat grnzK hum grnzA mus grnzA rat grnzM hum grnzM mus grnzM hum grnzB mus grnzB rat grnzB hum grnzH mus grnzC mus grnzD mus grnzE mus grnzF mus grnzG rat grnzJ hu mast chymase hu cathepsinG : : : : : : : : : : : : : : : : : : : : : NKIIKK----------KSNLVPPHTN------KSKLAPSRAH------KSKLAPSSAH------IMTIKGAV--------KKIMKGSV--------RKVIGRWSPQPLT---RKVTGRSA--------RKVIGRWSPQSLV---KKTMKRH---------KKTMKSS---------EETMKKS---------KRTMKRL---------KKTMKHS---------SWNMKLL---------SRNMKLL---------SRNMKLL---------STNMKLL---------QKTMKLLQQP------NQILQAN---------RTTMRSFKLLDQMETPL : : : : : : : : : : : : : : : : : : : : : 231 238 238 233 234 232 238 232 238 227 227 228 226 228 228 228 228 228 228 226 235 97 : : : : : : : : : : : : : : : : : : : : : 225 228 228 223 226 224 225 224 225 220 220 221 219 221 221 221 221 221 218 219 218 Figure 3.4. Phylogenetic analysis of catfish granzyme: Phylogram showing relationships of catfish granzyme to other known serine proteases. The tree was derived by parsimony analysis, with Mega version 2. Numbers shown above the branches are bootstrap values based upon 1000 replicates for parsimony. A separate analysis using maximum likelihood and neighbor joining methods produced a tree with similar topology. The tree was rooted on a sub-tree containing trypsin and chymotrypsin of fish and murine origin to determine the clustering of catfish granzyme with very similar known granzyme sequences. In addition to the previously described granzyme sequences, mature sequences of the following proteases were used to obtain the tree human adpsin (accession No. NP_001919), rat chymotrypsin B (accession No. AAA98732), rat pancreatic protease (accession No. NP_036861), lampray (Petromyzon marinus) trypsin (accession No. AAB69656), eel (Anguilla japonica) trypsin (accession No. BAB85634), and salmon (Salmo salar) trypsin (accession No. CAA49679). 98 99 Figure. 3.5. Analysis of tissue expression of catfish granzyme. (A) RT-PCR analysis was performed with catfish granzyme specific primers using cDNA from various tissues. Lane 1, Anterior Kidney; lane 2, Spleen; lane 3, Total Blood; lane 4, Liver; lane 5, Gill; lane 6, Muscle; lane 7, Trunk Kidney; lane 8, Heart; lane 9, NCC cDNA library (Positive Control). (B) NCC were enriched from three tissues to analyze the granzyme expression. Lane 1, NCC purified from peripheral blood, Lane 2, NCC purified from anterior kidney, Lane 3, NCC purified from spleen. 100 101 Figure 3.6. Alignment of other fish granzyme sequences with catfish granzyme. A, A portion of tilapia granzyme was amplified with primers used in cloning of catfish gene. The nucleotide sequence was edited, translated and aligned with catfish granzyme. Residues marked (•) above the alignment differ from catfish granzyme, but are highly similar to different mammalian granzymes. B, Partial sequence of a Salmo salar granzyme derived from an EST (Accession no CB516537) was edited and translated to compare with mature catfish granzyme. C, N-terminal region of a partial sequence from Salmo salar (accession no AF434669) which resembles human granzyme M was edited and translated to compare with the mature catfish granzyme. 102 (A) ccf grnz (CFGR1) : MHVQQRSLLLILTLFQATACSGSFIIGGREVKKPKPWMASVQSNNSHICGGTLIHQQWVL : ilapia grnz pcr : ------------------------------------------------------------ : 60 - ccf grnz (CFGR1) tilapia grnz pcr : TAAHCKTFLQFKPIEVLLGAHSLTKDKNAMRVKVLCFHISPKFSATTRVHDIMLLKLQDK : 120 : ---------------------------------------------LNFSNDIMLLKLSRK : 15 ccf grnz (CFGR1) tilapia grnz pcr : VQLKKNKVDVKKIPKSGKDIPAGTKCEVRGWGTTHVKNPKACDTLQELEVTVVDRELCNC : 180 : LQLDK-KVKPIQLARKEIKAKDNVKCQVAGWGFTETSG-KTVDVLRWVDVPLIDLNVCKR : 73 ccf grnz (CFGR1) tilapia grnz pcr : YYNSKPTITANMLCAGNKQRDKDACWGDSGGPLECKKNIVGVVSGGSGCGNPKKPGVYTL : 240 : KLKAKVTLPKGVVCAGGSDTKNGFCQGDSGGPLVCNN----------------------- : 110 ccf grnz (CFGR1) tilapia grnz pcr : LSKEHIDWINKIIKK : 255 : --------------- : - (B) CFGR-1 salmo est : ---------MHVQQRSLLLILTLFQATACSGSFIIGGREVK-KPKPWMASVQSNNSHICG : : ERFNRIIRFGGPLLQTALLIASVWKLAACSEVTIVGGHEVKPHSRPWMVSLQVKNNHVCG : 50 60 CFGR-1 salmo est : GTLIHQQWVLTAAHCKTFLQFKP--IEVLLGAHSLTKDKNAMRVKVLCFHISPKFSATTR : 108 : GTLIRDQWVLTAAHCKGVFGESKKFVEAVLGAHSLTSSKNTQRVGIEEYYVPGTYSDRTK : 120 CFGR-1 salmo est : VHDIMLLKLQDKVQLKKNKVDVKKIPKSGKDIPAGTKCEVRGWGTTHVKNPKACDTLQEL : 168 : EDDIMLIKLKMKVKINSKAVKVKEVSKSGKDLQVGTQCHVTGWGVVSATGSMPSDTLQGA : 180 CFGR-1 salmo est : EVTVVDRELCNCYYNSKPTITANMLCAGNKQRDKDACWGDSGGPLECKKNIVGVVSGGSG : 228 : EVDILDRKLCDCFYNRNPVITQDMLCASNNKRQADACKGDSGGPLECKKAFVGLVSGGLG : 240 CFGR-1 salmo est : CGNPKKPGVYTLLSKEHIDWINKIIKK : 255 : --------------------------- : - (C) CFGR-1 : MHVQQRSLLLILTLFQATACSGSFIIGGREVKKPKPWMASVQSNNSHICGGTLIHQQWVL : salmo grnzM-like : ----------MILSTAASFWTSTMILLYLHSGRGVCVCLCYPTNIYYYFSNEVTVGVLSL : 60 50 CFGR-1 : TAAHCKTFLQFKPIEVLLGAHSLTKDKNAMRVKVLCFHISPKFSATTRVHDIMLLKLQDK : 120 salmo grnzM-like : LLGDCAEIIGGKEVTPHSLPYMALLEDNKGNKKCGGILSHQQWVLTGAHC---------- : 100 CFGR-1 : VQLKKNKVDVKKIPKSGKDIPAGTKCEVRGWGTTHVKNPKACDTLQELEVTVVDRELCNC : 180 salmo grnzM-like : ------------------------------------------------------------ : CFGR-1 : YYNSKPTITANMLCAGNKQRDKDACWGDSGGPLECKKNIVGVVSGGSGCGNPKKPGVYTL : 240 salmo grnzM-like : ------------------------------------------------------------ : CFGR-1 : LSKEHIDWINKIIKK : 255 salmo grnzM-like : --------------- : - 103 Table 3.1. Oligonucleotide primers used in the cloning of catfish granzyme and for expression studies by RT-PCR. * AP (5' RACE Abridged Anchor Primer) and AUAP (Abridged Universal Amplification Primer) were obtained as a component of 5' RACE system (Invitrogen, CA). AP and AUAP are specifically engineered for the optimal amplification of dC-tailed cDNA. It contains strategically positioned deoxyinosine (I) and U residues to maximize specific priming at the dC-tailed end. Name Sequence Use F1 5’-GCTGCAGTAGCATGATGTCA-3’ Initial Screening R1 5’- GTTACACACAAGAGGGCCTCCA-3’ Initial Screening 5RC1 5’-GCATGATGTCATTGGAGAAGTTC-3’ 5' RACE 5RC2 5'-GGGAATCTTCTTAACGTCTAC-3' 5' RACE 5RC3 5'- GTTCCCCACCCTCTTACTTCAC-3' 5' RACE 3RC1 5'- CAAGCAGAGGGACAAAGATGCA-3' 3' RACE 3RC2 5'- TGGAGGCCCTCTTGTGTGTAAC-3' 3' RACE CDF1 5'-CCCGGATCCATGCACGTGCAACAACGTTC-3' ORF amplification CDR1 5'-GGGGTTCTAGATGCTTTTTAATTATCTTGTTGATCCAGTCA-3' ORF amplification EPB-18T 5'-GCGATTTCTGCAGGATCCAAACTT(17)-3' 3' RACE RT EPB 5'-GCGATTTCTGCAGGATCCAAACT-3' 3' RACE AP (*) 5'-CUACUACUACUACACGCGTCGACTAGTACGGGIIGGGIIGGGIIG-3' 5' RACE AUAP (*) 5'-CUACUACUACUACACGCGTCGACTAGTAC-3' 5' RACE GRRTF1 5'-GTTTCAAGCAACAGCTTGCTCAGGCA-3' RT PCR GRRTR1 5'-GCATAGGACTTTCACTCGCATAGCA-3' RT PCR ACTINF 5'-GACAATGGTTCCGGTATGTGCA-3' RT PCR ACTINR 5'-GGTGCCAGATCTTCTCCATGTCA-3' RT PCR 104 Table 3.2. Amino acid identity and similarity of mature catfish granzyme to other known granzymes and related proteins. Human GRNZ A Mouse GRNZ A Human GRNZ B Mouse GRNZ B Rat GRNZ B Mouse GRNZ C Mouse GRNZ D Mouse GRNZ E Mouse GRNZ F Mouse GRNZ G Human GRNZ H Rat GRNZ J Human GRNZ K Mouse GRNZ K Rat GRNZ K Human GRNZ M Mouse GRNZ M Rat GRNZ M Human Cathepsin G Human Mast Cell Chymase % Identity 41.0 39.7 32.1 35.9 37.0 30.3 31.6 30.3 31.1 32.4 32.9 34.6 43.3 45.8 45.4 33.1 32.5 33.9 31.2 37.4 105 % Similarity 53.1 53.2 40.9 46.8 46.6 41.2 43.0 42.0 42.4 43.7 43.9 45.4 55.4 55.8 55.0 47.5 42.9 47.1 44.5 49.2 CHAPTER 4 NONSPECIFIC CYTOTOXIC CELLS OF TELEOSTS ARE ARMED WITH MULTIPLE GRANZYMES AND OTHER COMPONENTS OF THE GRANULE EXOCYTOSIS PATHWAY1 1 Praveen, K., J. H. Leary, D. L. Evans and L. Jaso-Friedmann. To be submitted to Molecular Immunology after revision 106 ABSTRACT Granzymes are members of the serine protease family and major components of cytotoxic granules of professional killer cells. Multiple granzymes have been identified from human and rodents with different substrate specificities. Although the significance of granzymes A and B in cell-mediated cytotoxicity has been extensively investigated, recent reports suggest that other granzymes may have either equal or greater importance in mediating cell death. Studies on the evolution of these closely related proteases were hindered by the lack of sequence and biochemical information of granzymes from “lower vertebrates.” Here we report the generation of a catalytically active recombinant granzyme identified in the cytotoxic cells of an ectothermic vertebrate. Fully active, soluble recombinant catfish granzyme-1 (CFGR-1) was generated using a yeast-based expression system. In vitro enzyme kinetic assays using various thiobenzyl ester substrates verified its tryptase activity in full agreement with previous observations by sequence comparison and molecular modeling. The tryptase activity that was secreted from catfish NCC during an in vitro cytotoxicity assay strongly correlated with the cytotoxicity induced by these cells. Evidence for additional granzymes with different substrate specificities in NCC was obtained by analysis of the protease activity of supernatants collected from in vitro cytotoxicity assays. Searches of the catfish EST database further confirmed the presence of teleost granzymes with different substrate specificities. Granzyme activity measurements suggested a predominance of chymase and tryptase activities in NCC. Further proof that the granule exocytosis pathway is one of the cytotoxic mechanisms in NCC was provided by the expression of granule components perforin, granulysin and serglycin detected by RT-PCR analysis. These results demonstrate the evidence for a parallel evolution of effector molecules of cell-mediated cytotoxicity in teleosts. 107 Key words: Granzymes, perforin, granulysin, serglycin, thiobenzyl ester substrates, nonspecific cytotoxic cells INTRODUCTION Cell-mediated cytotoxicity against virally transformed and tumor cells is mediated through highly specialized lymphocytes. Cytotoxic T lyphocytes (CTLs) and natural killer cells (NK cells) are the most important among them (Lieberman, 2003). Although these two cells use distinct mechanisms to recognize their targets, following receptor engagement and activation they use the same effector pathways to mediate target cell death. One of the major effector mechanisms used by these cells, which would eventually lead to target cell death, involves release of perforin and a family of serine proteases called granzymes into the immunological synapse formed between an effector and its target cell (Podack, 1995, Wowk and Trapani, 2004). In addition to granzymes and perforin, other proteins are also present in the cytotoxic granules in smaller quantities, which appear to have supportive roles in killing by granule exocytosis (Smyth et al., 2001, Clark and Griffiths, 2003). Cytotoxic granules are considered secretory lysosomes because of the presence of lysosomal proteins along with lytic molecules, which can induce target cell death (Burkhardt et al., 1990, Peters et al., 1991). The granules have a morphologically distinct core consisting mainly of perforin and granzymes. The association of these lytic proteins with proteoglycans in a pH-dependant fashion, helps to maintain their enzymatic activity in an inactive state and thus, avoids damaging the effector cells (Masson et al., 1990). In fact, unlike conventional lysosomes which lack proteoglycans, the presence of serglycin in these granules, serves as a good indicator of the role of a cell in cell-mediated cytotoxicity (Clark and Griffiths, 2003). 108 The involvement of multiple granzymes in cell death pathways has been documented. Experimental expression of granzymes along with perforin in non-cytotoxic cells can make them efficient killers, suggesting that they are very important components of the granule exocytosis pathway of cytotoxicity (Hayes et al., 1989, Shiver et al., 1992). Experimental animals lacking select granzymes have been shown to suffer significant immunodeficiencies, although the phenotypic characteristics of the effects were focal. These results suggest that multiple granzymes, whose functions may not be clear at present, may be major players in death pathways (Trapani and Smyth, 2002, Ebnet et al., 1995, Wilharm et al., 1999b, Heusel et al., 1994, Revell et al., 2005). Granzymes are classified based on the four primary substrate specificities that have been described to date: tryptase (trypsin-like, cleaving after Arg/Lys), Asp-ase (similar to caspases, cleaving after Asp), Met-ase (cleaving after Met/Leu) and chymase (chymotrypsin-like, cleaving after Phe/Trp/Tyr) (Kam et al., 2000). The most extensively studied mammalian granzymes fall under three of the above specificities. Granzymes A and K have tryptase activity, granzyme B has Asp-ase activity, while granzyme M has Met-ase activity (Kam et al., 2000). The majority of the remaining granzymes, whose functions are not fully understood, have chymase specificity. Some of them have been implicated in the regulation of perforin activity (Hudig et al., 1993, Woodard et al., 1998). The significant roles that these chymases may be playing in novel cell death pathways are now widely appreciated. We have previously shown the presence of granzyme-like serine proteases in teleost nonspecific cytotoxic cells, NCC (Praveen et al., 2004). The first granzyme gene identified in catfish NCC was predicted to have a tryptase activity due to the presence of conserved triplet residues, which are important in the formation of the substrate specificity pocket (Wouters et al., 109 2003, Praveen et al., 2004). We now report on the expression of CFGR-1 as a recombinant protein and we show its specificity for Lys/Arg residues, in agreement with the previously predicted activity profile. The presence of other granzymes with different substrate specificities as well as additional components of cytotoxic granules is also reported. These results demonstrate the significance of NCC as a major cytotoxic cell population in channel catfish (Ictalurus punctatus). 2. MATERIALS AND METHODS 2.1. Production of mature and pro-CFGR-1 in Pichia pastoris The cDNAs coding for the pro and mature forms of CFGR-1 were PCR amplified and cloned into pPICZ-alpha vector for expression in GS115 strain of Pichia pastoris (Invitrogen, CA). Forward primers CFGRYSTF1 (5’-aactctcgagaaaagaatcataggaggccga-3’) and CFGRYSTF2 (5’-aactctcgagaaaagaagtttcatcataggaggccga-3’) were designed to engineer an Xho I site upstream of the mature and pro CFGR-1, respectively. A reverse primer CFGRYSTR1 (5’ggggttctagatgtgttcctttgagagaagagtatagac-3’) was designed to incorporate an Xba I site. Combinations of these primers were used to PCR amplify the insert using a full-length CFGR-1 clone, which was previously sequenced in both directions (Praveen et al., 2004). The PCR products were purified, digested with restriction enzymes (Xho I and Xba I) and ligated into the similarly cut pPICZ-alpha vector. The insert was introduced into this vector downstream of the alpha factor signal sequence and Kex2 protease cleavage site. The insert’s position facilitates the production of recombinant proteins with a native N-terminus and a polyhistidine tag at the Cterminus as well as secretion to the medium with minimum contamination from yeast proteins. As is the case with other yeast expression systems, the vector was grown in E. coli before 110 expression in yeast cells. Thus, the ligation reaction was first used to transform competent JM109 E. coli cells and positive clones were selected on a low-salt LB plate with 25 µg/ml Zeocin (Invitrogen, CA). The clones were verified for the presence of insert by restriction digestion and colony PCR methods. The two chosen clones (pPICZ-alpha-CFGR-1 and pPICZalpha-proCFGR-1) were sequenced in both directions using 5’ and 3’ AOX1 primers (Invitrogen, CA) to verify the correct reading frame. The recombinant plasmids were linearized by digesting with Sac I before electroporation into GS115 strain of Pichia pastoris according manufacturer’s instructions. Positive clones were selected on YPD agar plates containing 100 µg/ml Zeocin. Positive clones were tested for insert by PCR and expression of recombinant proteins were assessed by small scale expression trial followed by Western blot analysis of TCA precipitated supernatants. Clones with high level of expression were selected for scale-up. In order to obtain enough recombinant protein for purification, the selected clones were grown in baffled flasks for optimum aeration. Using a single colony, 25 ml of buffered glycerolcomplex medium [BMGY: 1% yeast extract, 2% peptone, 100 mM potassium phosphate (pH = 6.0), 1.34% yeast nitrogen base, 4 x 10-5 % biotin, 1% glycerol] in a 250 ml baffled flask was incubated at 30o C for 18 hours until the OD600 reached 2-6. Cells were harvested by centrifugation and pellet was resuspended in buffered methanol-complex medium [BMMY: BMGY with glycerol replaced with 0.5% methanol] to an OD600 of 1.0 in a 1 liter baffled flask. The flasks were incubated at 30o C for various time points, till the optimum production was verified by Western blotting of supernatants collected at different time points. The supernatants were collected and stored at -80o C until ready for purification. His-tagged recombinant protein purification was accomplished by concentration of the supernatants with ammonium sulfate precipitation, dialysis in phosphate buffered saline (PBS, 111 pH = 7.2), followed by metal affinity chromatography using Ni-NTA agarose (Qiagen, CA) according to manufacturer’s instructions. The positive fractions were pooled, desalted and concentrated using Centricon YM-3 centrifugal filters (Millipore, MA). Aliquots were stored at 80o C. Protein concentrations were estimated using Biorad Protein Assay Kit (Biorad, CA). 2.2. Western blotting Proteins were resolved on 12.5% gel, transferred to nitrocellulose membrane and probed with INDIA His probe-HRP (Pierce, IL) or anti-His monoclonal antibody (Qiagen, CA). Proteins were detected with SuperSignal West Pico Chemiluminescent Substrate (Pierce, IL). 2.3. In vitro enzyme assays Activity assays for proteases were performed at 37o C in 50 mM Tris-HCl, 0.15 M NaCl, 0.01% Triton X-100 (pH = 7.6) containing 0.2 mM Ellman’s reagent [5,5’-dithiobis(2nitrobenzoic acid), Sigma, MO] in 96-well microtiter plates with a reaction volume of 200 µl. Various thiobenzyl ester substrates at a concentration of 0.2 mM were used to determine the enzyme kinetics by measuring increase of absorbance at 405 nm wavelength over time with a Spectra Max Plus microplate reader (Molecular Devices, CA). Results for controls and samples are expressed as change in mOD/min. Stock solutions of Z-Lys-SBzl (Calbiochem, CA) were made in ethanol, while Z-Arg-SBzl, Boc-Ala-Ala-Asp-SBzl and Boc-Ala-Ala-Met-SBzl (MP Biomedicals, OH) were dissolved in dimethyl sulfoxide (DMSO) and stored at -20o C. Stock solution of substrates were added to the reaction buffer prior to the assay. 112 2.4. Granule exocytosis NCC were prepared from anterior kidney of channel catfish as described before (Praveen et al., 2004). The target cells (HL-60) were obtained from American Type Culture Collection (ATCC, VA) and were grown as suspension cultures in RPMI-1640 supplemented with 10% heat-inactivated FBS. Cells were washed three times in PBS containing 5% BSA and diluted to 1×105 cells/ml. Targets (10,000 cells in 100 µl volume) were delivered into 96-well round bottom microtiter plates. NCC were added (in 100 µl) at different concentrations to attain several E:T ratios and the plates were centrifuged (500 x g) to allow the formation of effector-target cells soft pellets. After incubation for 4 hours, the plates were centrifuged again at 1200 x g and supernatants were collected for measurement of protease activity. 2.5. Cell-mediated Cytotoxicity Target cells (2×106 HL-60 cells/ml) were labeled with 20 µCi of sodium chromate (Amersham, IL) for 3 h at 37°C. Cells were washed three times in medium and diluted to 1×105 cells/ml. Labeled targets were mixed with effector cells (catfish NCC) to attain different E:T ratios and the plates were centrifuged at 500 x g. After a 4 hour incubation, 100 µl of each supernatant was removed to determine radioactivity (Cobra II Autogamma counter; Packard, CT). The results are expressed as percentage specific release (SR). Percent SR = (Test Release – Spontaneous Release/ Total Release – Spontaneous release) ×100. 2.6 Identification of cytotoxic granule components in catfish NCC Comparisons of full-length mammalian perforin sequences with the EST sequences for channel catfish in NCBI identified three overlapping ESTs (CB939050, CB937333 and 113 CB937672) with considerable sequence similarity to N-terminal, central and C-terminal regions of mammalian perforins. These EST sequences were edited, and joined together to obtain the full open reading frame for catfish perforin. Primers were designed based on this sequence and used in nested RT-PCR of cDNA from catfish NCC isolated from anterior kidney. The primers used were: outer primers CFPERFF1: 5’-cctggctcaagaaggacaagtcct-3’ and CFPERFR1: 5’ccacccagagtcacctttgagatat-3’, nested primers CFPERFF2: 5’-gcgaccaaatcagaagtgcagaac-3’ and CFPERFR2: 5’-gcgaaaccctgtatctgtagtatcca-3’. Similarly, ESTs with similarity to known granulysin (CB938093) and serglycin (CK411555) sequences were identified and used to design primers (CFGRYF1: 5’-gctcgttgcttctttcttcataggc-3’, CFGRYR1: 5’-gcgaattgtgctttaatcatgtcca-3’ and CFSRGF1: 5’-cggataatgtactcggagcctca-3’, CFSRGR1: 5’-gggttacgttccttccggctta-3’) to amplify those molecules by RT-PCR. 3. RESULTS 3.1. Expression of recombinant CFGR-1 in Pichia pastoris We previously identified the first non-mammalian cDNA sequence from a granzyme (CFGR-1) from the cytotoxic cells of a teleost fish (Praveen et al., 2004). Comparison of this serine protease with other granzymes from human and rodents suggested a tryptase activity for CFGR-1. In order to assess the primary substrate specificity for this protease, we expressed it as a recombinant protein in Pichia pastoris. Selection of the expression system was based on the assumption that this protease could be expressed in eukaryotic cells in its native form without the need for refolding. The coding region of CFGR-1 was amplified and cloned in between Xho I and Xba I sites of the expression vector pPICZ-alpha. For pro-CFGR-1, two amino acids upstream of the N-terminus of the mature protein (Ser and Phe) were retained by making the 114 appropriate changes in the forward primer used to amplify the insert. Introduction of these amplicons at this position resulted in an in-frame fusion between Sacharomyces cervisiae alpha factor mating signal sequence and pro/mature CFGR-1 (Fig 1a). PCR primers were engineered to incorporate a Kex2 cleavage site in between the signal sequence and CFGR-1. This facilitated the processing of the signal sequence by Pichia so that mature/pro CFGR-1 with native Nterminus was released into the culture supernatants. At the C-terminus of the recombinant protein, a c-myc epitope and poly histidine tag were incorporated to enable efficient purification and detection of the proteins (Fig. 1a). Previous studies have demonstrated that small C-terminal tags can be incorporated in proteases without affecting the proper folding and activity (Beresford et al., 1997, Huang et al., 1997, Huang et al., 1998, Xia et al., 1998). In pPICZ-alpha, the expression cassette is under the influence of highly-inducible AOX1 promoter, which can drive the expression of the recombinant protein with methanol induction. Several clones were selected from YPD-Zeocin plates and expressions of recombinant proteins were assessed by growing in 10 ml BMGY medium in 50 ml conical tubes. Clones with highest level of expression were selected for further studies. Selected clones were grown and induced to secrete the recombinant proteins in 300 ml culture volume in a 1000 ml baffled flasks and culture supernatants were collected at various time points for TCA precipitation. Later, the proteins were resolved on a 12.5% SDS-PAGE and transferred to nitrocellulose. Expression levels of recombinant proteins were assessed by staining for His-probe. A protein of approximately 33 kDa was found to be upregulated by methanol in both mature and pro CFGR-1 culture supernatants. For mature CFGR-1, significant level of expression was noticed only after 72 hours of induction. In case of pro-CFGR-1, high level of expression was observed as early as 24 hours. Prolonged incubation resulted in degradation of the proteins in both instances. 115 Optimum time of induction for large scale production was selected based on these results (Fig. 1b). 3.2. Large scale production and purification of recombinant CFGR-1 Ni-NTA agarose beads were mixed with a small volume (20 ml) of the culture supernatants and His-tagged proteins were purified to assess the success of metal affinity chromatography. Almost all of the recombinant proteins were eluted in the first three elutions, detected by Western blotting and protease activity assays (data not shown). Based on these results, large volumes (1-2 liters) of the culture supernatants were subjected to ammonium sulfate precipitation to concentrate the proteins. Nickel affinity chromatography was used to purify the His-tagged proteins (Fig. 2). 3.3. In vitro proteolytic activity of CFGR-1 Comparison of CFGR-1 amino acid sequences with other mammalian granzymes suggested the primary specificity determining triplet residues as DGG, similar to that in granzymes A and K. Therefore, Z-lys-SBzl was selected to check the proteolytic activity of the purified CFGR-1. As expected from the sequence comparisons and molecular modeling studies (Praveen et al., 2004), CFGR-1 hydrolyzed the thiobenzyl ester designed to study tryptase-like proteases. The pro-CFGR-1 had no activity suggesting the retaining the two pro-peptides at the N-terminus of the protease can affect the proper folding and activity of CFGR-1 (Fig 3a). The most preferred P1 residues for CFGR-1 was determined to be Arg and Lys, similar to that of granzyme A and K (Fig 3b). Mature CFGR-1 lacked the Met-ase or Asp-ase activity as determined by the enzyme kinetics assays using Boc-Ala-Ala-Met-SBzl and Boc-Ala-Ala-Asp- 116 SBzl substrates, respectively (Fig 3b). In all these cases, pro-CFGR-1 did not exhibit proteolytic activities. 3.4. Correlation between the NCC cytotoxicity and exocytosis of tryptases The enzymatic activity of supernatants from NCC and target cells were collected for protease activity measurement and correlated with Cr release cytotoxicity assays. Catfish NCC were mixed with target cells (HL-60) at different E:T ratios in duplicate experiments. In one set of experiments, the targets were labeled with 20 µCi of sodium chromate in RPMI with 10% fetal bovine serum. In the other set of experiments, nonlabeled target cells were incubated with NCC in PBS with 10% BSA, in order to minimize the effects of anti-protease activity of serum on the protease assays. Cytotoxicity was determined by measuring the radioactivity in the supernatants of the first set of experiments and granule exocytosis was determined by measuring the hydrolysis of Z-Lys-SBzl by the supernatants collected from the second set of experiments. There was a strong correlation between the cytotoxicity and residual tryptase activity in the supernatants (Fig 4). These results suggested the presence of preformed tryptases like CFGR-1 in the cytotoxic granules of NCC. 3.5. Granzyme-like protease activities in catfish NCC granules The presence of other serine proteases with activities similar to mammalian granzyme was determined by measuring the Asp-ase, Met-ase, tryptase and chymase activities in the supernatants after incubating NCC with target cells for four hours. The highest enzymatic activity in the supernatants was chymase, followed by tryptase, while little Asp-ase and Met-ase 117 activities were detected (Fig 5). The possibility of the contribution to enzymatic activity from target cells was ruled out by measuring the protease activities of total cell lysates of HL-60. 3.6. Expression of additional granzyme-like proteases in NCC. Although the proteolytic activities in supernatants from NCC contained at least two substrate specificities, this alone was not sufficient evidence for the presence of other granzymes in NCC. Other cDNA sequences containing signature granzyme motifs were searched in the EST databases. Partial sequences for various proteases expressed in various hematopoietic tissues were identified. Among them, three sequences had motifs characteristic of all granzymes identified so far. The comparison of deduced amino acid sequences for these proteins with CFGR-1 suggested conservation of residues around the catalytic triad and also the N-terminal region (Fig 6a). However, the triplet residues, which determine the primary substrate specificity varied considerably from CFGR-1, suggesting deviation from tryptase activity for these proteases (Fig 6a). The three proteins were arbitrarily named as CFGR-2, CFGR-3 and CFGR-4 for identification. CFGR-3 and 4 are peculiar due to the fact that they have Gly at position 186 (chymotrypsin numbering), which is not seen in any other granzyme sequences. Another teleost granzyme identified from tilapia (Praveen et al., 2004) also had similar triplet residues and was shown to have a chymase activity (Praveen et al. Manuscript in preparation). Based on these findings, at least two of the catfish granzymes identified can be predicted to have chymase activity. Expression of these granzyme sequences in catfish NCC was analyzed by RT-PCR. Catfish NCC express all the four granzymes identified so far (Fig 6b). This might explain the 118 high level of chymase activity observed in the supernatants when NCC were mixed with target cells. 3.7. Expression of cytotoxic granule-associated proteins in NCC. Expression of granule-associated proteins like perforin, granulysin, serglycin etc was analyzed by RT-PCR. By searching catfish EST database using sequences for similar proteins from mammalian sources as query, partial sequences spanning various regions of such genes were obtained. Using that sequence information, primers were designed to amplify portions of those genes. Intron spanning regions (by comparing the mammalian gene sequences for the corresponding proteins) were selected for primer design to make sure a different size product is amplified while cDNA or genomic DNA is used as template. Catfish NCC was shown to express perforin, granulysin and serglycin cDNAs (Fig 7), indicating the presence of all major granule components. The identity of the amplicons was verified by sequencing and it was found to correspond to the catfish homologs of the indicated proteins. 4. DISCUSSION The ability to express purified granzymes in recombinant form is crucial in understanding their individual role in various physiological processes. Purification of catalytically active mammalian granzyme B, using yeast expression system has been previously reported (Pham et al., 1998, Sun et al., 1999). Similarly, other expression systems like mammalian cells (Smyth et al., 1995) and insect cells with recombinant baculovirus (Xia et al., 1998) were successfully used to generate active granzyme B. Attempts to express granzyme B in E. coli as a fusion protein with maltose binding protein was not successful because of the inability of the protein to fold 119 correctly and regain solubility (Sun et al., 1999). However, catalytically active recombinant granzyme A and K could be expressed in E. coli (Beresford et al., 1997, Wilharm et al., 1999a). The granzymes were first generated as a pro-enzyme and processed with enterokinase or bovine cathepsin C after refolding of the precursor to regain the activity. Successful expression of recombinant human granzyme A zymogen in many mammalian cell lines was reported after infection with recombinant vaccinia virus (Kummer et al., 1996). Expressing the protease as an inactive zymogen allowed elimination of proteolytic damage to the expression system. However, the need for further processing of these pro-enzymes necessitated extra steps in the purification process. Granzyme M also was expressed successfully in mammalian cells using CDM8 mammalian expression vector (Kelly et al., 1996). Other similar expression trials for related proteases include the use of Pichia pastoris for human mast cell tryptase (Niles et al., 1998) human mast cell chymase (Lockhart et al., 2005), insect cells for mouse mast cell protease 6 and 7 (Huang et al., 1998, Huang et al., 1997). Properly engineered amino terminus starting with intact I (I/V)GG motif has been shown to be essential for the granzymes to adopt an active conformation. Here, expression cassette for the mature CFGR-1 was engineered by amplifying the portion of the gene coding for amino acids staring at Ile-25 of the pre-pro enzyme (CFGR with propeptides and signal sequence intact). Similarly, pro-CFGR-1 was generated by retaining two amino acids at the N-terminus (Ser-23 and Phe-24). The presence of this pro-peptide was intended for preventing the proper folding of the enzyme, providing a negative control for the activity assays, as it has been demonstrated for many other proteases (Pham et al., 1998, Sun et al., 1999). The use of pPICZalpha vector facilitated the secretion of the recombinant proteins in to the supernatants due to the presence of an alpha mating signal sequence upstream of the gene of interest. Presence of Kex2 120 cleavage site between the signal sequence and CFGR-1 insert allowed the generation of the recombinant proteins with a desired N-terminal end, as compared to the addition of vectorencoded residues at the N-terminus of recombinant proteins expressed with other vectors (Fig 1a). The supernatants were collected at various time points to check the expression level of the recombinant proteins by Western blot analysis. Addition of two small tags at the C-terminus provided means of easy detection either with antibodies against c-myc/poly-histidine (data not shown) or with INDIA-His probe (Fig 1b). Once the conditions were optimized, the production of recombinant proteins was scaledup and supernatants were collected for protein purification by nickel affinity chromatography. (Fig. 2). In vitro protease assays using thiobenzyl esters demonstrated that the mature CFGR-1 was active even in the presence of imidazole used in the elution of the Ni-NTA agarose columns (data not shown). As it had been demonstrated for other proteases, the presence of two amino acids at the N-terminus prevented the proper folding of the recombinant CFGR-1, which is evident from the lack of activity for pro-CFGR-1 (Fig 3a). Comparison of crystal structures of granzymes and other serine proteases has lead to the theory that three residues of S1 substrate binding pocket confer primary specificity to these proteases. These residues are located at position 189, 216 and 226 (chymotrypsin numbering) (Shotton and Hartley, 1970, Perona and Craik, 1997, Tsu et al., 1997, Wouters et al., 2003). By comparing the CFGR-1 sequence with other proteases, this granzyme had been previously predicted to have a tryptase activity due to the S1 specificity pocket residues (DGG) similar to granzyme A and K (Praveen et al., 2004). Using recombinant CFGR-1 and thiobenzyl ester substrates designed for tryptase, Asp-ase and Met-ase, the fine specificity of this granzyme towards basic residues (Arg or Lys) at P1 was identified (Fig 3b). 121 The initial description of catfish NCC was as agranular lymphocytes because of the lack of large cytotoxic granules in these small cells by Giemsa staining and light microscopy (Evans et al., 1984). However, their dependence on the presence of calcium for cytotoxic activity was an indication that these cells might utilize perforin and granzymes to induce cell death (Carlson et al., 1985). Subsequently, the molecular evidence for the expression of granzyme-like proteases in these cells indicated that NCC could contain very small granules, difficult to visualize by normal microscopic techniques (Praveen et al., 2004). CFGR-1 was identified as one of the main tryptase like granzymes found in catfish NCC. Correlation between the exocytosis of proteases and the cytotoxicity induced by these cells was determined by measuring target cell death by chromium release assay along with the tryptase activity of supernatants. The strong correlation between the cytotoxicity and the tryptase activity suggested CFGR-1 is released on target cells by NCC (Fig. 4). However, these observations cannot provide the exact contribution of CFGR-1 towards the cytotoxicity. Conjugate formation between NCC and target cells can be expected to result in release of effector molecules in to the immunological synapse. Some of it will be taken up by the target cell and the rest will remain in the supernatants. Measuring the protease activities of the supernatants using the thibenzyl ester substrates designed for common granzymes will give biochemical evidence for the types of granzymes exocytosed by the effector cells during conjugate formation. Catfish NCC had pre-formed granzymes stored inside, which was released towards the target cells upon conjugate formation. The main activity observed was chymase and tryptase, but very little Met-ase and Asp-ase activity (Fig. 5). However, this would not guarantee the existence of granzymes with novel substrate specificities than the existing mammalian granzymes. 122 Molecular evidence for the expression of multiple granzymes in catfish NCC came from the RT-PCR studies using primers designed based on EST sequences (Fig. 6b). Comparison of predicted amino acid sequences for these granzymes supported the findings from the granule exocytosis assay (Fig 6a). Only CFGR-1 had a specificity pocket triplet similar to granzyme A and K (DGG). All the remaining granzymes do have the triplet similar to known chymases. The closest similarity of these sequences was with another teleost granzyme identified in tilapia cytotoxic cells (Praveen et al., 2004). This granzyme has been expressed and its substrate specificity has been verified (Praveen et al. Manuscript in preparation). Following a similar strategy, expression of other granule-associated proteins was also assessed in catfish NCC (Fig 7). Of particular interest was to determine if NCC had proteoglycans (such as serglycin), known to be specific for cytotoxic cells. Our observations suggest that cytotoxic cells of teleosts have multiple effector molecules of the granule exocytosis pathway, similar to mammalian cytotoxic lymphocytes. The localization of these molecules in cytoplasmic granules can only be verified by immuno-electron microscopic studies using antibodies specific for fish granular proteins. These reagents are presently not available. In conclusion, catfish NCC were found to express components of the granule exocytosis pathway of cell-mediated cytotoxicity. Based on the mammalian serine protease evolution, CFGR-1, which has tryptase activity, can be predicted to have co-evolved with other granzymes with more specific substrate specificities. These studies predict a parallel evolution of cellmediated cytotoxicity in ectothermic vertebrates. Future studies are aimed at determining the physiological role played by individual granzymes in induction of cell death or regulation of NCC functions. 123 ACKNOWLEDGEMENTS We acknowledge the help of Dr. Dorothy Hudig and Dr. Dieter Jenne for their valuable suggestions and providing reagents and expertise. This work was funded with support from USDA (98-35205-6701). REFERENCES Beresford, P.J., Kam, C.M., Powers, J.C., Lieberman, J., 1997. Recombinant human granzyme A binds to two putative HLA-associated proteins and cleaves one of them. Proc. Natl. Acad. Sci. U. S. A 94, 9285-9290. Burkhardt, J.K., Hester, S., Lapham, C.K., Argon, Y., 1990. The lytic granules of natural killer cells are dual-function organelles combining secretory and pre-lysosomal compartments. J. Cell Biol. 111, 2327-2340. Carlson, R.L., Evans, D.L., Graves, S.S., 1985. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). V. Metabolic requirements of lysis. Dev. Comp Immunol. 9, 271-280. Clark, R., Griffiths, G.M., 2003. Lytic granules, secretory lysosomes and disease. Curr. Opin. Immunol. 15, 516-521. Ebnet, K., Hausmann, M., Lehmann-Grube, F., Mullbacher, A., Kopf, M., Lamers, M., Simon, M.M., 1995. Granzyme A-deficient mice retain potent cell-mediated cytotoxicity. EMBO J. 14, 4230-4239. Evans, D.L., Carlson, R.L., Graves, S.S., Hogan, K.T., 1984. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). IV. Target cell binding and recycling capacity. Dev. Comp Immunol. 8, 823-833. Hayes, M.P., Berrebi, G.A., Henkart, P.A., 1989. Induction of target cell DNA release by the cytotoxic T lymphocyte granule protease granzyme A. J. Exp. Med. 170, 933-946. Heusel, J.W., Wesselschmidt, R.L., Shresta, S., Russell, J.H., Ley, T.J., 1994. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76, 977-987. Huang, C., Friend, D.S., Qiu, W.T., Wong, G.W., Morales, G., Hunt, J., Stevens, R.L., 1998. Induction of a selective and persistent extravasation of neutrophils into the peritoneal cavity by tryptase mouse mast cell protease 6. J. Immunol. 160, 1910-1919. Huang, C., Wong, G.W., Ghildyal, N., Gurish, M.F., Sali, A., Matsumoto, R., Qiu, W.T., Stevens, R.L., 1997. The tryptase, mouse mast cell protease 7, exhibits anticoagulant 124 activity in vivo and in vitro due to its ability to degrade fibrinogen in the presence of the diverse array of protease inhibitors in plasma. J. Biol. Chem. 272, 31885-31893. Hudig, D., Ewoldt, G.R., Woodard, S.L., 1993. Proteases and lymphocyte cytotoxic killing mechanisms. Curr. Opin. Immunol. 5, 90-96. Kam, C.M., Hudig, D., Powers, J.C., 2000. Granzymes (lymphocyte serine proteases): characterization with natural and synthetic substrates and inhibitors. Biochim. Biophys. Acta 1477, 307-323. Kelly, J.M., O' Connor, M.D., Hulett, M.D., Thia, K.Y., Smyth, M.J., 1996. Cloning and expression of the recombinant mouse natural killer cell granzyme Met-ase-1. Immunogenetics 44, 340-350. Kummer, J.A., Kamp, A.M., Citarella, F., Horrevoets, A.J., Hack, C.E., 1996. Expression of human recombinant granzyme A zymogen and its activation by the cysteine proteinase cathepsin C. J. Biol. Chem. 271, 9281-9286. Lieberman, J., 2003. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nat. Rev. Immunol. 3, 361-370. Lockhart, B.E., Vencill, J.R., Felix, C.M., Johnson, D.A., 2005. Recombinant human mast-cell chymase: an improved procedure for expression in Pichia pastoris and purification of the highly active enzyme. Biotechnol. Appl. Biochem. 41, 89-95. Masson, D., Peters, P.J., Geuze, H.J., Borst, J., Tschopp, J., 1990. Interaction of chondroitin sulfate with perforin and granzymes of cytolytic T-cells is dependent on pH. Biochemistry 29, 11229-11235. Niles, A.L., Maffitt, M., Haak-Frendscho, M., Wheeless, C.J., Johnson, D.A., 1998. Recombinant human mast cell tryptase beta: stable expression in Pichia pastoris and purification of fully active enzyme. Biotechnol. Appl. Biochem. 28 ( Pt 2), 125-131. Perona, J.J., Craik, C.S., 1997. Evolutionary divergence of substrate specificity within the chymotrypsin-like serine protease fold. J. Biol. Chem. 272, 29987-29990. Peters, P.J., Borst, J., Oorschot, V., Fukuda, M., Krahenbuhl, O., Tschopp, J., Slot, J.W., Geuze, H.J., 1991. Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and granzymes. J. Exp. Med. 173, 1099-1109. Pham, C.T., Thomas, D.A., Mercer, J.D., Ley, T.J., 1998. Production of fully active recombinant murine granzyme B in yeast. J. Biol. Chem. 273, 1629-1633. Podack, E.R., 1995. Functional significance of two cytolytic pathways of cytotoxic T lymphocytes. J. Leukoc. Biol. 57, 548-552. Praveen, K., Evans, D.L., Jaso-Friedmann, L., 2004. Evidence for the existence of granzyme-like serine proteases in teleost cytotoxic cells. J. Mol. Evol. 58, 449-459. 125 Revell, P.A., Grossman, W.J., Thomas, D.A., Cao, X., Behl, R., Ratner, J.A., Lu, Z.H., Ley, T.J., 2005. Granzyme B and the Downstream Granzymes C and/or F Are Important for Cytotoxic Lymphocyte Functions. J. Immunol. 174, 2124-2131. Shiver, J.W., Su, L., Henkart, P.A., 1992. Cytotoxicity with target DNA breakdown by rat basophilic leukemia cells expressing both cytolysin and granzyme A. Cell 71, 315-322. Shotton, D.M., Hartley, B.S., 1970. Amino-acid sequence of porcine pancreatic elastase and its homologies with other serine proteinases. Nature 225, 802-806. Smyth, M.J., Kelly, J.M., Sutton, V.R., Davis, J.E., Browne, K.A., Sayers, T.J., Trapani, J.A., 2001. Unlocking the secrets of cytotoxic granule proteins. J. Leukoc. Biol. 70, 18-29. Smyth, M.J., McGuire, M.J., Thia, K.Y., 1995. Expression of recombinant human granzyme B. A processing and activation role for dipeptidyl peptidase I. J. Immunol. 154, 6299-6305. Sun, J., Bird, C.H., Buzza, M.S., McKee, K.E., Whisstock, J.C., Bird, P.I., 1999. Expression and purification of recombinant human granzyme B from Pichia pastoris. Biochem. Biophys. Res. Commun. 261, 251-255. Trapani, J.A., Smyth, M.J., 2002. Functional significance of the perforin/granzyme cell death pathway. Nat. Rev. Immunol. 2, 735-747. Tsu, C.A., Perona, J.J., Fletterick, R.J., Craik, C.S., 1997. Structural basis for the broad substrate specificity of fiddler crab collagenolytic serine protease 1. Biochemistry 36, 5393-5401. Wilharm, E., Parry, M.A., Friebel, R., Tschesche, H., Matschiner, G., Sommerhoff, C.P., Jenne, D.E., 1999a. Generation of catalytically active granzyme K from Escherichia coli inclusion bodies and identification of efficient granzyme K inhibitors in human plasma. J. Biol. Chem. 274, 27331-27337. Wilharm, E., Tschopp, J., Jenne, D.E., 1999b. Biological activities of granzyme K are conserved in the mouse and account for residual Z-Lys-SBzl activity in granzyme A-deficient mice. FEBS Lett. 459, 139-142. Woodard, S.L., Fraser, S.A., Winkler, U., Jackson, D.S., Kam, C.M., Powers, J.C., Hudig, D., 1998. Purification and characterization of lymphocyte chymase I, a granzyme implicated in perforin-mediated lysis. J. Immunol. 160, 4988-4993. Wouters, M.A., Liu, K., Riek, P., Husain, A., 2003. A despecialization step underlying evolution of a family of serine proteases. Mol. Cell 12, 343-354. Wowk, M.E., Trapani, J.A., 2004. Cytotoxic activity of the lymphocyte toxin granzyme B. Microbes. Infect. 6, 752-758. Xia, Z., Kam, C.M., Huang, C., Powers, J.C., Mandle, R.J., Stevens, R.L., Lieberman, J., 1998. Expression and purification of enzymatically active recombinant granzyme B in a baculovirus system. Biochem. Biophys. Res. Commun. 243, 384-389. 126 Figure 4.1. Production of active and pro-CFGR-1 in Pichia pastoris. (a) Schematic representation of the expression cassettes constructed to express the recombinant proteins using plasmid pPICZ-alpha. The recombinant proteins were expressed as fusion proteins starting with the yeast alpha factor at the N-terminus and poly-histidine and c-myc tagged granzyme at the Cterminus. Presence of a Kex2 site in between the alpha factor and the granzyme allowed the expression of granzyme with a native N-terminus, which can be secreted in to the culture medium with minimum contaminating yeast proteins. (b) Optimization of recombinant protein expression. The culture supernatants were collected at the time points indicated and subjected to TCA precipitation followed by Western blotting. His-tagged proteins were detected with INDIAHis probe conjugated to HRP followed by chemiluminescence. Yeast trsansfected with empty vector alone (E) also were checked for the expression of proteins (only 72 hour time point is shown). 127 A B 128 Figure 4.2. Purification of recombinant proteins. Supernatants were collected after methanol induction for 72 hours and total proteins were concentrated by ammonium sulfate precipitation. His-tagged proteins were purified by metal affinity chromatography. Purification success was verified by Western blotting and detection with INDIA-His probe. Lane V: vector alone, lane F: flow though from the column, lane E1, E2, and E3: three consecutive elusions of the column using buffer containing 250 mM imidazole. 129 130 Figure 4.3. Enzymatic activity of recombinant CFGR-1. (a) Plot depicting the change in optical density vs. time from an in vitro enzyme kinetic assay using Z-lys-SBzl as a substrate. (b) Substrate specificity of CFGR-1. The enzymatic activity of mature and pro-CFGR-1 was measured using the indicated thiobenzyl ester substrates in final concentrations of 0.3 mM. Enzyme concentration was approximately 1 nM for both mature and pro-CFGR-1. 131 A B Boc-Ala-Ala-Asp-Sbzl Boc-Ala-Ala-Met-Sbzl CFGR-1 Pro-CFGR-1 Z-Arg-SBzl Empty vector Z-Lys-SBzl 0 1 2 3 4 Change in mOD/min 132 5 6 Figure 4.4. Correlation between cytotoxicity and exocytosis of granzymes with tryptase activity in catfish NCC. Radioisotope labeled HL-60 targets were incubated with catfish NCC at various effector:target ratios for four hours and cytotoxicity was measured by determining the residual radioactivity in the supernatants. In a parallel experiment, nonlabeled targets were incubated with effector cells and the supernatants were collected to measure the residual tryptase activity by hydrolysis of Z-Lys-SBzl. 133 7 35 6 30 5 25 4 20 3 15 2 10 5 1 0 0 160 80 40 E:T Ratio 134 20 Change in mOD/min % Specific Release 40 Cytotoxicity Protease Activity Figure 4.5. Expression of other proteases in the cytotoxic granules of catfish NCC. HL-60 target cells were incubated with NCC for four hours at various E:T ratios and supernatants were collected to measure the residual protease activity using different thiobenzyl esters to determine various protease activities associated with known granzymes (Z-Lys-SBzl for tryptase, NSuccinyl-Phe-Leu-Phe-SBzl for chymase, Boc-Ala-Ala-Asp-SBzl for Asp-ase and Boc-Ala-AlaMet-SBzl for Met-ase). The spontaneous release of any granzymes from HL-60 was also determined for each kind of activity. Also, the target cells were lysed to determine the presence of any granzyme activity from those cells. 135 Metase Aspase HL-60 lysate HL-60 Only Tryptase NCC + HL-60 Chymase 0 10 20 30 Change in mOD/min 136 40 50 Figure 4.6. Identification of multiple granzymes from catfish NCC. (a) Three additional granzyme-like proteases sequences from channel catfish were identified by searching the EST database. Multiple sequence alignment of predicted amino acid sequences for those proteases with CFGR-1 shows conserved motifs, characteristic of known granzymes. The residues at the relevant positions in the specificity pocket triplet, are represented with small circles above the alignment. (b) RT-PCR analysis of cDNA from catfish head kidney NCC for the expression of multiple granzymes. Control reactions were run using genomic DNA as a template with primers that would result in a product with bigger size. This ensured that the source of the amplicons was cDNA and not contaminating genomic DNA. 137 A CGRR-1 CFGR-2 CFGR-3 CFGR-4 : : : : IIGGREVKK-PKPWMASVQSNNSHICGGTLIHQQWVLTAAHCKTFLQFKP---IE IIGGNEVDRHSRPYMASVQFKKAHMCGGFLIRKDYVLTAAHCVDNIDHSGKDKLE IINGKKAKKNSFQYMASVQSKGKHICGGFLISPSYVLTAAHCFQSN-------LS IVNGSVVTANSRPYMVSVQKDGKHLCGGFLMSENFVMTAAHCWEAG-----VKLT : : : : 51 55 48 50 CGRR-1 CFGR-2 CFGR-3 CFGR-4 : : : : VLLGAHSLTKDKNAMR-VKVLCFHISPKFSAT-TRVHDIMLLKLQDKVQLKKNKV VLLGAHNINQKESQQQRIQVQKYILHPCYERG-ERPNDIMLLKLKSKAKENKF-V VVLGTQNIDAKRNELRRYAVKSMHIHPSYKENPRYGSDIMLLKFSGKVNLNKD-L VVVGSHELKKSKSAISHMEVKLYHIHPKFDSE-NLLNDIMLLQLKKTIKKSKN-I : : : : 104 108 102 103 CGRR-1 CFGR-2 CFGR-3 CFGR-4 : : : : DVKKIPKSG-KDIPAGTKCEVRGWGTTHVKNPKACDTLQELEVTVVDRELCNCYY KVIALPKKD-ENLPARQECSIAGWGKTKQ-NSAESSVLREVKLKLENNSQCKKFW KVIKISSNH-KRVKPNTKCQVAGWGKT-E-TQKTVNDLMVTDVSTIDITVCKKQW NWISIPKKMNQDVKAKQVCSIAGWGKKSE-NGGTSDRLMEVNVTVIDTKVCEKSW : : : : 158 161 154 157 CGRR-1 CFGR-2 CFGR-3 CFGR-4 : : : : NS-KPTITANMLCAGNKQRDKDACWGDSGGPLECKKNIVGVVSGGS--GCGNPKK Q--NYFDTDNMICTVSD-GKKAFCQGDSGSPLICGNEPQGIAAYTHPHDCLNPTY NKENVELPAKILCAGGYGTKSGACQGDSGGPLVCSGLAVGIVSFNLHDNCSYPNV G--TPFSVSSLVCTK---GRGGFCQGDSGGPLVCQNTAVGIVSFTD-ADCKYPKN : : : : 210 213 209 206 CGRR-1 CFGR-2 CFGR-3 CFGR-4 : : : : PGVYTLLSKEHIDWINKIIKK--PGVYMKIS-YFLPWIKQVIH---PNVYTEIS-AYADWINKVIKRDAPYVYTKIS-TFSALD--------- : : : : 231 232 231 220 B 138 Figure 4.7. Expression of other granule components in catfish NCC. RT-PCR analysis of cDNA isolated from catfish NCC using primers designed based on EST sequences similar to mammalian granule-associated proteins. (a) Expression of perforin in catfish NCC. Lane Lib: cDNA library, cDNA: fresh cDNA, gDNA: catfish genomic DNA, NTC: no template control (b) Nested PCR for perforin, using cDNA as a template (c) Expression of granulysin in catfish NCC. (d) Expression of serglycin in catfish NCC. 139 A B C D 140 CHAPTER 5 MOLECULAR CHARACTERIZATION AND EXPRESSION OF A GRANZYME WITH CHYMASE ACTIVITY FROM CYTOTOXIC CELLS OF AN ECTOTHERMIC VERTEBRATE1 1 Praveen, K., J. H. Leary, D. L. Evans and L. Jaso-Friedmann. To be submitted to Journal of Immunology after revisions 141 ABSTRACT We have identified the gene coding for a novel serine protease with close similarities to mammalian granzymes from nonspecific cytotoxic cells of a teleost fish Oreochromis niloticus. The genomic organization of tilapia granzyme-1 (TLGR-1) has the signature five exon-four intron structure shared by all granzymes. Several sequence motifs are shared among the teleost and mammalian granzymes, including the N-terminal tetrapeptides, catalytic triad residues constituting the charge relay system and residues surrounding the triad. Molecular modeling studies suggested a granzyme-like structure for this protein with four disulfide linkages and two additional Cys residues. The expression of this gene is found to be restricted to cytotoxic cell populations with a low level of constitutive expression when compared to similar granzymes in other teleost species. High levels of transcriptional activation of TLGR-1 with different stimuli suggested that this gene is highly induced during immune reactions. Triplet residues around the active site Ser of TLGR, which determine the primary substrate specificity of granzymes, differ significantly from that of other granzymes. Recombinant TLGR-1 was expressed in mature or pro-enzyme forms using pPICZ-alpha vector in Pichia pastoris expression system. Recombinant TLGR-1 was used to determine the primary substrate specificity of this protease using various synthetic thiobenzyl ester substrates. In vitro enzyme kinetics assays suggested a preference for residues with bulky side chains at the P1 site, indicating a chymase-like activity for this protease. These results indicate the presence of novel granzymes in cytotoxic cells from ectothermic vertebrates. Key words: Granzyme, chymase, thiobenzyl ester, nonspecific cytotoxic cells, lymphocytes, serine proteases, cell-mediated cytotoxicity 142 INTRODUCTION Granzymes belong to a family of serine proteases, which constitute the major components of granules of professional killer cells such as cytotoxic T lymphocytes (CTLs) and natural killer cells (NK cells). Along with perforin, a pore forming protein that is also found in these granules, granzymes play a major role in inducing target cell death (1-3). All the mammalian granzymes described so far are highly conserved and are located in three clusters on three chromosomes (4). Granzymes A and B are the most extensively studied granzymes. The different pathways used by granzymes A and B to initiate cytotoxicity have been described (5,6). However, novel mechanisms of cytotoxicity are being discovered for these granzymes, expanding our knowledge about complex pathways involved in cell death and proliferation (712). The function of many of the other granzymes is not well understood, thus they are collectively called as orphan granzymes (4). Non-redundant pathways of cell-mediated cytotoxicity have been proposed for granzymes, suggesting a ‘fail-safe’ mechanism provided by multiple granzymes (2,4). The role of granzymes in inducing target cell death has been investigated extensively in vitro (13). However, advances in in vivo studies demonstrating the significance of individual granzymes in cytotoxicity lag behind. Targeted gene disruption studies of granzymes have resulted in focal immune deficiencies (13-16). Recent attempts to understand the individual effects of members of the granzyme B gene cluster has revealed the significant role played by orphan granzymes present downstream of the granzyme B gene in mice (17). Such results indicate the relative significance of other granzymes that have not been fully investigated. Granzymes are classified based on their primary substrate specificities and four enzymatic activities have been described: tryptase , Asp-ase, Met-ase and chymase. The most 143 extensively studied mammalian granzymes fall under three of the above specificities. Granzyme A and K have tryptase activity, granzyme B has Asp-ase activity, while granzyme M has Met-ase activity (18). Many of the orphan granzymes have chymase activity. The presence of multiple granzymes can be interpreted as a result of molecular strategies developed by the immune system to compete with pathogens. These pathogens have coexisted and coevolved with immune systems of organisms for millions of years (13). Genetic comparisons and chromosomal clustering of granzymes suggest gene duplication events as a means of generating newer granzymes (4). Almost all the assumptions on granzyme evolution are based on the gene sequence information from human and rodents, due to the lack of sequence information in other organisms. The availability of non-mammalian granzyme sequences would add new insights into the evolution of these proteases with very narrow substrate specificity and it would also aid in the discovery of newer killing pathways (19,20). Nonspecific cytotoxic cells (NCC) are the first identified cytotoxic cell population in teleosts. First described in the channel catfish (21), these cells have been well characterized in a number of lower vertebrates (22-25). Although first described as agranular (21), NCC in trout, tilapia and catfish do have small granules and contact with target cells leads to granule exocytosis with necrotic and apoptotic death pathways (26-28). The requirement for calcium for the induction of cytotoxicity by NCC was an early indication that these cells use the granule exocytosis killing pathway (29). We have previously reported the existence of granzyme-like serine proteases in NCC of channel catfish (Ictalurus punctatus) and tilapia (Oreochromis niloticus) (20). Here, evidence is presented of the full-length sequence of the cDNA and gene for TLGR-1, its expression pattern and the substrate specificity of the recombinant protein. These 144 results demonstrate for the first time, the molecular characterization of granzymes from a cytotoxic cell of non-mammalian vertebrates with novel substrate specificities. MATERIALS AND METHODS Cloning of full-length TLGR-1 The identification and partial sequence of a granzyme-like serine protease from a tilapia NCC cDNA library has been previously reported (20). Full-length sequence of that tilapia granzyme (TLGR-1) was obtained by rapid amplification of cDNA ends (RACE). Fresh RNA was purified from tilapia NCC from peripheral blood for the RACE, as previously described (20). Briefly, mRNA (500 ng) was reverse transcribed with granzyme specific primers using Generacer Superscript II RT module (Invitrogen, CA). First strand cDNA was subjected to homopolymeric tailing with cytosine using terminal transferase (Roche). Later, dC-tailed cDNA was amplified using abridged anchor primers and gene specific nested primers followed by a reamplification using a nested abridged universal amplification primer (AUAP) and another nested gene-specific primer. The amplicons were purified and TA cloned for sequencing. For 3'RACE, RNA was reverse transcribed using an anchor primer, EPB-18T (30) to generate first strand cDNA. Using gene specific nested primers and EPB, the 3'end of the mRNA was amplified and TA cloned for sequencing. The sequences were edited and assembled to obtain the complete sequence of TLGR-1. Forward and reverse primers were designed at the two ends to amplify the full-length cDNA, which was TA cloned and sequenced in both directions using standard protocols. 145 TLGR-1 gene and promoter sequences The complete genomic sequence of TLGR-1 gene and its promoter elements were obtained by sequencing a cosmid clone containing the TLGR-1 gene. The tilapia genomic DNA library was constructed in SuperCos-1, as reported for channel catfish (31). A directed PCRbased iterative screening protocol was used to identify clones with TLGR-1 gene (32). Specific PCR primers (TLGRCOSF1 5’-gcacccacgatatgacaaagttga-3’ and TLGRCOSR1 5’ggcctttcaattttctcttacagaca-3’) were designed based on the cDNA sequence. After several rounds of screening, several TLGR-1 positive clones were isolated, expanded and frozen in glycerol stocks for further analysis. The complete TLGR-1 gene sequence was obtained by sequencing the cosmid clone in a 373 A DNA sequencer (Applied Biosystems, CA) at the Integrated Biotech Laboratories (University of Georgia, Athens). The upstream region of the TLGR-1 gene (promoter region: 1200 bp) was directly sequenced from the cosmid. Primers were synthesized as needed for “primer walking" (Genosys, TX). The TRANSFAC database (33) and MatInspector web based software (34) were used to locate transcription factor binding sites. Phylogenetic analysis and molecular modeling Phylogenetic analysis and molecular modeling of TLGR-1 were done as described before (20). Briefly, all the known granzymes and similar serine proteases were aligned and phylogenetic analysis was done with Mega version 2.1 (35). Analysis using different methods resulted in similar trees and reliability of the trees was tested using 1000 bootstrap replications. The tree was rooted on a subtree consisting of adipsin-like proteases from various sources. Molecular modeling of TLGR-1 was done using SWISS-MODEL in the first approach mode 146 accessible via the internet (http://www.expasy.org/swissmod). RasWin version 2.6 was used to visualize the coordinate data. Real-time PCR analysis After exposing NCC activation treatments, total RNA was isolated using RNeasy mini-kit (Qiagen, CA) following the manufacturer’s protocol, quantified spectrophotometrically and treated with RNAse free DNAse (Promega, WI). The first strand cDNA was synthesized from 3 µg of total RNA using First Strand cDNA synthesis kit (Invitrogen, CA). A cDNA stock for constructing a relative standard curve was synthesized using the same method. Primers were designed based on available gene sequences to amplify ~150 bp amplicons for TLGR-1 and betaactin. Forward and reverse primers were selected at positions to include an intron so that genomic DNA contamination in the samples could be easily detected. A relative standard curve was constructed for target gene and house-keeping gene ( -actin) using either cDNA stock or full length cDNA clone for TLGR-1 and -actin. Briefly, reactions were set-up with different concentrations ranging from 10 to 3000 pg per reaction for the standard curve. The reactions were carried out using Brilliant® SYBR Green QPCR master mix (Stratagene, CA) according to manufacturer’s instructions. The efficiencies of the primer pairs were determined and appropriate adjustments were made to optimize reaction conditions. Diluted cDNA samples were used as a template for every 25 µl reaction. Each sample was set-up in triplicates for the PCR. The following PCR protocol was used for gene amplification: 95 °C, 3 min; 40 cycles: denature 95 °C, 30 s; anneal 60 °C, 30 s; extend 72 °C, 30 s, followed by 4 °C hold using Mx3000P realtime PCR system (Stratagene, CA). 147 Production of mature and pro-TLGR-1 in Pichia pastoris The cDNAs coding for pro and mature forms of TLGR-1 were PCR amplified and cloned in to pPICZ-alpha vector for expression in GS115 strain of Pichia pastoris (Invitrogen, CA). Forward primers TLGRYSTF1 (5’-aactgtcgacaaaagaatcataaatggca-3’) and TLGRYSTF2 (5’aactgtcgacaaaagaagtgaaatcataaatggca-3’) were designed to engineer an Xho I site upstream of mature and pro TLGR-1 respectively. The coding region of TLGR-1 contained a recognition site for Xba I. Therefore, in order to clone the insert between Xho I and Xba I sites of pPICZ-alpha, a reverse primer TLGRYSTR1 (5’-ggggttctagatggcattgctttttattgagaatgt-3’) was designed to incorporate a Sal I site. Digestion of DNA with Sal I has been shown to generate cohesive ends compatible with that made with Xba I digestion. Combination of these primers were used to PCR amplify the insert using a full-length TLGR-1 clone, which was previously sequenced in both directions. The PCR products were purified and digested with a combination of restriction enzymes (Xho I and Sal I) and ligated in to the pPICZ-alpha vector digested with Xho I and Xba I. The insert was introduced into this vector downstream of alpha factor signal sequence and Kex2 protease cleavage site. This position facilitated the production of recombinant proteins with native N-terminus and a polyhistidine tag at the C-terminus, which would be secreted to the medium with minimum contamination from yeast proteins. The ligation reaction was used to initially transform competent JM109 E. coli cells and positive clones were selected on a low-salt LB plate with 25 µg/ml Zeocin (Invitrogen, CA). The clones were verified for the presence of insert by restriction digestion and colony PCR methods. Recombinant plasmids (pPICZ-alphaTLGR-1 and pPICZ-alpha-proTLGR-1) were sequenced in both directions using 5’ and 3’ AOX1 primers (Invitrogen, CA) to verify the correct reading frame. Subsequently, the recombinant plasmids were linearized by digesting with Sac I before electroporating in to GS115 148 strain of Pichia pastoris according manufacturer’s instructions. Positive clones were selected on YPD agar plates containing 100 µg/ml Zeocin. Positive clones were tested for insert by PCR. Expression of recombinant proteins was assessed by a small scale expression trial followed by Western blot analysis of the TCA precipitated supernatants. Clones with high level of expression were selected to scale-up production. In order to obtain enough recombinant protein for purification, the selected clones were grown in baffled flasks for optimum aeration. Using a single colony, 25 ml of buffered glycerolcomplex medium [BMGY: 1% yeast extract, 2% peptone, 100 mM potassium phosphate (pH = 6.0), 1.34% yeast nitrogen base, 4 x 10-5 % biotin, 1% glycerol] in a 250 ml baffled flask was incubated at 30o C for 18 hours until the OD600 = 2-6. Cells were harvested by centrifugation and the pellet was resuspended in buffered methanol-complex medium [BMMY: BMGY with glycerol replaced with 0.5% methanol] to an OD600 of 1.0 in a 1 liter baffled flask. The flasks were incubated at 30o C for various time points, till the optimum production was verified by Western blotting of supernatants collected at different time points. The supernatants were collected and stored at -80o C until ready for purification. For purification of the His-tagged recombinant proteins, the supernatants were concentrated by ammonium sulfate precipitation followed by dialysis in phosphate buffered saline (PBS, pH = 7.2). The recombinant proteins were purified by metal affinity chromatography using Ni-NTA agarose (Qiagen, CA) according to manufacturer’s instructions. The positive fractions were pooled, desalted and concentrated using Centricon YM-3 centrifugal filters (Millipore, MA). Aliquots were stored at -80o C. Protein concentrations were estimated using Biorad Protein Assay Kit (Biorad, CA). 149 Western blot analysis Proteins were resolved on 12.5% gel, transferred to nitrocellulose membrane and probed with INDIA His probe-HRP (Pierce, IL) or anti-His monoclonal antibody (Qiagen, CA). Proteins were detected with SuperSignal West Pico Chemiluminescent Substrate (Pierce, IL). In vitro enzyme assays Activity assays for proteases were performed at 37o C in 50 mM Tris-HCl, 0.15 M NaCl, 0.01% Triton X-100 (pH = 7.6) containing 0.2 mM Ellman’s reagent [5,5’-dithiobis(2nitrobenzoic acid), Sigma, MO] in 96-well microtiter plates with a reaction volume of 200 µl. Various thiobenzyl ester substrates at a concentration of 0.2 mM were used to determine the enzyme kinetics by measuring increase in absorbance at 405 nm wavelength over time with a Spectra Max Plus microplate reader (Molecular Devices, CA). Results for controls and samples are expressed as change in mOD/min. Stock solutions of Z-Lys-SBzl (Calbiochem, CA) were made in ethanol, while Z-Arg-SBzl, Boc-Ala-Ala-Asp-SBzl, Boc-Ala-Ala-Met-SBzl (MP Biomedicals, OH), N-Succinyl-Phe-Leu-Phe-SBzl and N-Succinyl-Ala-Ala-Pro-Phe-pNA (Sigma, MO) were dissolved dimethyl sulfoxide (DMSO) and stored at -20o C and added to the reaction buffer prior to the assay. RESULTS Identification and analysis of full-length cDNA and gene for TLGR-1 For identification of genes involved in cytotoxic functions of NCC, a cDNA library was constructed using activated cytotoxic cells. The identification of a partial sequence of a serine protease with novel predicted substrate specificity from tilapia NCC library has been reported 150 (20). Later, full-length cDNA for that gene was identified and sequenced. Primers were designed based on this sequence to screen a tilapia genomic library and a cosmid clone containing the gene and promoter elements was identified (Fig. 1). Surprisingly, the TLGR-1 gene had an approximate size of 1.32 kb (starting from the 5’ UTR and ending at the polyadenylation signal), which is much smaller than mammalian granzyme genes. However, comparison of the cDNA and genomic sequences of TLGR-1 gene, revealed a five exon and four intron organization, shared among many members of the granzyme family (Table 1). The teleost granzyme gene has shorter introns than the mammalian granzyme genes. The promoter region of TLGR-1 has transcription factor binding sites, which are similar to other granzymes. All of the intron/exon boundaries of TLGR-1 gene fulfill the GT-AG rule (36). The proximal promoter region of TLGR-1 was analyzed for the putative transcription factor binding sites. Similar to other granzyme gene promoters, TLGR-1 has many binding sites for transcription factors specific for the immune system (Fig. 1). There are two AP-1 and NFAT binding sites along with sites for binding of interferon response factors and STATs. This would appear to suggest that there may be a significant correlation between the transcriptional upregulation of TLGR-1 and activated immune response reactions. TLGR-1 cDNA sequence was initially obtained by RT-PCR using specific primers at the 5’ and 3’ end of the gene and the sequence was verified by comparing it with the genomic sequence. The cDNA and genomic sequences were submitted to GenBank (accession numbers AY918866 and AY918867, respectively). This cDNA has a total length of 999 bp with an open reading frame of 765 bp that encodes a putative protein with 254 amino acids (Fig. 1). Using algorithms to predict signal sequences, a stretch of 23 residues at the N-terminal end was identified as a signal sequence, indicating that this protein is sorted to the secretory pathway. 151 Signal sequence cleavage sites were predicted using weight matrix analysis of von Heijne (37). TLGR-1 is predicted to have a dipeptide (Ser-Glu) as pro-peptide, much similar to the majority of mammalian granzymes. There is a single putative glycosylation site (NGTA) located towards the C-terminus of the protein (Fig. 1). The predicted molecular mass of the non-glycosylated mature TLGR-1 was calculated as 25,339 daltons (229 amino acids) with an isoelectric point of 9.57. Other granzymes also have a highly basic charge. Disulfide linkages are crucial for the the proper folding of fully active granzymes. Molecular modeling revealed that the overall structure of tilapia granzyme had a close similarity to available crystal structures of mammalian granzymes. TLGR-1 can be predicted to have four disulfide linkages (Fig. 2), similar to what was shown for catfish granzyme (20). Formation of disulfide bonds between Cys residues can be predicted as follows: 42 with 58; 136 with 168; 182 with 201. Cys191 and Cys220 (chymotrypsinogen numbering) could be analogous to a fourth disulfide linkage which bridges active site serine, like in chymotrypsin. Although the tilapia granzyme has additional Cys residues (Fig. 2), these are not present at corresponding positions to that of granzyme A, which is crucial in the dimerization (19). Multiple sequence comparisons of TLGR-1 with related granzyme sequences enabled the identification of signature motifs shared by members of the granzyme family. While the Nterminal IIXG motif is conserved in TLGR-1, the PHSRPYMA motif is not entirely conserved. However, the residues in corresponding position share close similarity to those in the mammalian sequences. The three key amino acid residues representing the catalytic triad (charge relay system) of serine proteases (His57, Asp102 and Ser 195: chymotrypsinogen numbering system) as well as their neighboring residues are well conserved in TLGR-1 (Fig. 3). The combination of amino acid residues, which are crucial in the formation of the substrate specificity pocket, can be 152 used as a prediction tool for primary specificity for a protease (38). Based on comparison of the residues in TLGR-1, alignment with other granzyme sequences and manually editing the alignment based on crystallography structure profile (39), a novel substrate specificity pocket triplet (GNN) can be predicted for this teleost granzyme. None of the other granzymes described to date share a specificity pocket triplet with TLGR-1. TLGR-1 has overall sequence similarity with almost all of the mammalian granzymes, with highest sequence similarity to granzyme B and G sequences (Table 2). Phylogenetic analysis of TLGR-1 TLGR-1 sequence was compared with known serine proteases and the data was analyzed by different methods to obtain clues on evolutionary position of this granzyme. Phylogenetic analysis using different methods yielded trees with similar topology and a representative tree is depicted in Fig 4. In order to resolve the difference between closely related granzyme sequences, the tree was rooted on a subtree comprising of complement factor D, which has been shown to be evolutionarily distinct from granzymes while sharing many sequence features. TLGR-1 didn’t cluster with any of the mammalian granzymes. However, it grouped with other teleost granzymes identified by EST database searches. Comparison of substrate specificity pocket triplet residues in the teleost sequences revealed a close similarity to TLGR-1 suggesting that they may share similar primary specificities. Expression pattern of TLGR-1 Expression of TLGR-1 gene in various tissues was assessed by RT-PCR analysis using specific primers. Purified NCC were shown to be a major tissue for expression of this gene (Fig. 153 5). Tissues rich in NCC were the only places where the expression could be detected. Compared to the granzymes from catfish NCC, constitutive expression of TLGR-1 was found to be very low, based on the intensity of the amplified bands. Transcriptional regulation of TLGR-1 Inducibility of the TLGR-1 message in tilapia NCC was assessed by gene quantification with real-time RT-PCR. NCC were treated with various stimuli and cDNA were prepared for analysis. Recombinant tilapia TNF was found to have a profound effect on the transcription of TLGR-1 (Fig. 6a). Within two hours of treatment with 100 ng/ml recombinant tilapia TNF, there was a 29 fold increase in TLGR-1 expression. The expression was reduced after two hours, but remained higher than base levels even after four hours. Similarly, treatment of NCC with PMA/calcium ionophores resulted in more than two fold increase in TLGR-1 expression (Fig 6b). More than six fold increase in expression could be observed by treating the cells with LPS for four hours (Fig 6c). Expression of recombinant TLGR-1 In order to express the pro and mature forms of TLGR-1, expression cassettes were engineered so that it could be inserted between Xho I and Xba I sites of yeast expression vector, pPICZ-alpha. Recombinant vectors were introduced in to GS115 strain of Pichia pastoris so that the recombinant proteins were expressed as a fusion protein with an N-terminal alpha signal sequence and C-terminal granzyme. By inserting a Kex2 recognition sequence in between the signal sequence and granzyme, active (cleaved) TLGR-1 could be secreted in to the culture supernatant. Incorporation of small C-terminal tags without affecting the protease activity has 154 been demonstrated for many proteases before (40-43). Both pro and mature recombinant TLGR1 had a His-tag followed by a c-myc tag at the C-terminus (Fig. 7a). Expression of pro-TLGR-1 was achieved by retaining the activation dipeptide at the N-terminus, which is known to yield inactive enzyme due to improper protein folding. Several expression clones were identified by plating the transformed cells on YPD plates with Zeocin. Small scale expression trials were done to assess the level of expression of selected clones upon induction with methanol (data not shown). Clones with highest expression levels were selected for further studies. Single yeast colony was grown and induced to secrete the recombinant proteins in 300 ml culture volume in a 1000 ml baffled flasks and culture supernatants were collected at various time points for TCA precipitation. The proteins were resolved on a 12.5% SDS-PAGE and transferred to nitrocellulose. Expression levels of recombinant proteins were assessed by staining with His-probe. A protein of approximately 33 kDa was found to be upregulated in both mature and pro TLGR-1 culture supernatants (Fig. 7b). For mature TLGR-1 high level of expression of recombinant proteins were observed from 24 hours lasting till 72 hours. From that point time on, there was significant degradation of the proteins. Similarly, expression levels were high for pro-TLGR-1 from 48 hours and remained high for 96 hours (Fig. 7b). High level expression and purification of recombinant TLGR-1 In order to assess the success of metal affinity chromatography, Ni-NTA agarose beads were mixed with a small volume (20 ml) of the culture supernatants and His-tagged proteins were purified eluting the bound proteins in a buffer containing 250 mM imidazole. Almost all of the recombinant proteins were eluted in the first elutions, detected by Western blotting and 155 protease activity assays (data not shown). Later, large volumes (1-2 liters) of the culture supernatants were subjected to ammonium sulfate precipitation to concentrate the proteins. This was followed by nickel affinity chromatography to purify His-tagged proteins (Fig. 8). In vitro proteolytic activity of recombinant TLGR-1 Due to the novel specificity pocket triplet residues in TLGR-1, it was expected not to hydrolyze thiobenzyl esters specially designed for granzymes with tryptase, Met-ase or Asp-ase activity. The hydrolysis of synthetic substrates used to study proteases with chymase specificity was tried to analyze the activity of TLGR-1. Effective hydrolysis of N-Succinyl-Phe-Leu-PheSBzl (Fig 9a) and N-Succinyl-Ala-Ala-Pro-Phe-pNA (Fig 9b) by TLGR-1 suggested its strong preference for bulky residues at P1 site. Later, a comparison of hydrolysis of various thiobenzyl ester substrates revealed in fact, that TLGR-1 had chymase activity TLGR-1 (Fig 10). The proTLGR-1 failed to cleave any of the substrates, indicating that the presence of pro-peptide at the N-terminus prevented proper folding of the protease which is crucial for its activity. DISCUSSION We have previously reported the expression of granzyme-like serine proteases in NCC purified from peripheral blood of tilapia (20). The full-length cDNA coding for TLGR-1 was identified and sequenced in two directions to obtain the putative amino acid sequence of the protein and to identify sequence motifs shared with other granzymes. Later, a genomic library was screened using specific primers to obtain a cosmid clone containing full-length TLGR-1 gene. We sequenced genomic DNA comprising the full gene and the promoter region (Fig. 1). Compared to other granzyme sequences, TLGR-1 gene had a smaller size. However, the intron- 156 exon structure of the gene had closest similarity to other mammalian granzymes (1,44). Additionally, the number and position of introns of TLGR-1 suggests that this could be a member of the sixth class of serine proteases (45), like NK cell specific granzyme M (44,46). The first one of the substrate specificity triplet residues (Gly 173; -6 with reference to active site Ser) is encoded within the fourth exon (Fig 1 and Table 1) as in the case of other chymotrypsinlike proteases (47). Studies on proximal promoter elements of mammalian granzymes had revealed the conserved sequences enabling cell-specific expression of these proteases. Granzyme B promoter has binding sites for T cell specific transcription factors such as Ikaros and core-binding factor as well as for more ubiquitous transcription factors like AP-1 (48-50). These sequences are found to be sufficient to drive reporter gene expression in T cell lines where these factors are constitutively active (51,52). Promoter region of granzyme M gene has been shown to have binding sites for factors like AP-2, AP-3 and glucagons-G3A, influencing its bias for expression in NK cells (44,53). The presence of multiple transcription factors in the promoter region of the TLGR-1 gene in NCC is an indirect indication that this gene is highly regulated during the immune response. Highly conserved Cys residues at similar positions in all granzyme sequences give rise to three pairs of disulfide bonds, which are crucial in proper folding and activation of granzyme during processing. Among these disulfide bonds, the ones between Cys26 and Cys42 as well as between Cys151 and Cys167 link the primary and secondary substrate-binding sites, forming S1 subsites in granzymes A, K and M (54). A fourth pair of Cys residues form an additional disulfide bond, which bridges the active site Ser residue is found in granzymes M and K (44,55). Granzyme A is the only mammalian granzyme shown to have one additional Cys residue, which 157 is responsible for intermolecular disulfide bonds (19,56). TLGR-1 was shown to have two additional Cys residues (Fig 2). At this time their function is unknown as their positions do not match with those corresponding to the dimer-forming Cys residue of granzyme A (19). Multiple sequence alignment of TLGR-1 with other granzymes revealed highly conserved regions shared by all known granzymes (Fig. 3). The N-terminal IIXG motif and catalytic triad residues that make up the charge relay system were highly conserved. Moreover, residues around the triad residues were highly conserved among all the sequences. The presence of Gly residue at position -6 upstream of the active site Ser indicated a substrate specificity that was different from that of granzymes A/K, B and M. Other residues at position +15 to +17 and +28 relative to the active site Ser are found to be important in determining the primary substrate specificity of Ser protease (57). TLGR-1 has Ser-Phe-Asn (+15 to +17) and Asn (+28) at these positions, which is quite different from all other known granzymes. A comparison of crystal structures of Ser proteases has led to a theory that three residues of the S1 substrate binding pocket confer primary specificity. These specificity-conferring residues are located at positions 189, 216, and 226 (chymotrypsin numbering) (38). Alignment of the TLGR-1 sequence with other granzymes and matching the secondary structure elements, the substrate specificity pocket residues for this protease were determined as Gly-Asn-Asn. These triplet amino acid residues have not been previously described in mammalian granzymes. Multiple sequence comparisons alone are not sufficient to draw conclusions on evolutionary positions of closely related proteins like granzymes. Phylogenetic analysis of mammalian granzymes has revealed the clustering of granzymes with similar substrate specificity, giving rise to the classification of granzymes in to three major groups (4,38). Similar analysis of teleost granzymes along with mammalian serine protease placed TLGR-1 in to a 158 separate cluster, indicating a parallel evolution for teleost granzymes. Catfish granzymes with similar predicted substrate specificity were the other members of that cluster. The experimental verification of the enzymatic activity for the catfish granzymes would be crucial to make the correlation between the substrate specificity and sequence similarity. The pattern of tissue expression of TLGR-1 suggests that this protease is associated with cytotoxic cell populations (Fig. 5). Tissues with appreciable TLGR-1 expression were the ones with high NCC density. Use of cDNA derived from total tissues for expression studies could be misleading due to the interference from different cell populations that might be present in these tissues. The cell line (TMB-8), which was derived from heart tissue, was used to assess the specificity of expression with more confidence. TMB-8 cells have been used as nonhematopoietic and non-cytotoxic controls in previous experiments (58). Compared to the CFGR1, which had a high level of constitutive expression in NCC from different tissues (20), TLGR-1 expression levels were very low in resting NCC (Fig. 5). Transcriptional upregulation of TLGR1 with various stimuli indicated the high degree of inducibility for this tilapia granzyme (Fig. 6). Transcriptional activation of granzyme A in response to TNF has been previously reported in lymphokine-activated killer cells and CTLs (59,60). Purification of catalytically active recombinant granzymes is critical in understanding their individual physiological roles, compared to combined effects of multiple granzymes in purified cytotoxic granules. Generation of mammalian granzymes in recombinant forms have been reported (40,43,61-65). Use of Pichia pastoris expression system with pPICZ-alpha expression vector allowed the generation of recombinant granzymes with desired N-terminal end. By inserting the tilapia granzyme expression sequence downstream of an alpha mating signal sequence, secretion of the protein to the expression medium could be achieved. The pro- 159 TLGR-1 was generated by retaining two residues at the N-terminus of the mature protease (Fig 7a). This has been shown to prevent the proper folding and activation of mammalian recombinant granzymes (62,65,66). The supernatants were collected at various time points and expression levels were determined by Western blotting (Fig. 7b) Purification of recombinant TLGR-1 from culture supernatants was carried out after concentrating the proteins by ammonium sulfate precipitation. This method had been previously used without affecting the proteolytic activities of human mast cell chymase (67). After the ammonium sulfate precipitation, the pellet was dissolved in PBS and concentrated to 50 ml followed by dialyzing against PBS to remove traces of ammonium sulfate. His-tagged proteins were purified by nickel affinity chromatography (Fig. 8). The Western blot analysis of TLGR-1 suggests that the protein might be glycosylated. The predicated molecular mass of mature TLGR-1 is 25.3 kDa, while the protein appeared to have an approximate mass of 33 kDa, suggesting the presence of sugar residues in the predicted glycosylation site on the polypeptide chain. Glycosylation of granzymes is critical because they have been shown to be targeted to the lytic granules by the mannose-6-phosphate receptor (68). Granzymes are characterized by their very narrow substrate specificity. The requirement for P1 residue varies significantly between different classes of mammalian granzymes (18). In most cases the substrate specificity of individual granzymes can be predicted based on the architecture of its substrate binding pocket (38). Such prediction algorithms suggested that TLGR-1 had a preference for bulkier P1 residues than that for tryptases, Asp-ases or Met-ases. Using synthetic substrates designed with a bulky Phe residue at P1 site, the activity of TLGR-1 was shown to be similar to that of mammalian chymases (Fig. 9 & 10). 160 In conclusion, the novel serine protease identified from tilapia NCC was shown to have chymase activity. It is not clear whether these cells do express other granzymes and other components of granule exocytosis pathway. The TLGR-1 gene is highly inducible with various stimuli. Compared to the expression pattern of similar granzymes in channel catfish NCC, tilapia granzymes require longer activation for the transcription and translation of cytotoxic molecules, as determined by the protease activity of supernatants from granule exocytosis assay (data not shown). Studies are underway to uncover the physiological roles of this protease in these cytotoxic cells and also to identify other granule components in tilapia NCC. REFERENCES 1. Trapani, J. A. 2001. Granzymes: a family of lymphocyte granule serine proteases. Genome Biol. 2: Reviews 3014. 2. Lieberman, J. 2003. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nat. Rev. Immunol. 3:361. 3. Russell, J. H., and T. J. Ley. 2002. Lymphocyte-mediated cytotoxicity. Annu. Rev. Immunol. 20:323. 4. Grossman, W. J., P. A. Revell, Z. H. Lu, H. Johnson, A. J. Bredemeyer, and T. J. Ley. 2003. The orphan granzymes of humans and mice. Curr. Opin. Immunol. 15:544. 5. Trapani, J. A., and V. R. Sutton. 2003. Granzyme B: pro-apoptotic, antiviral and antitumor functions. Curr. Opin. Immunol. 15:533. 6. Lieberman, J., and Z. Fan. 2003. Nuclear war: the granzyme A-bomb. Curr. Opin. Immunol. 15:553. 7. Beresford, P. J., D. Zhang, D. Y. Oh, Z. Fan, E. L. Greer, M. L. Russo, M. Jaju, and J. Lieberman. 2001. Granzyme A activates an endoplasmic reticulum-associated caspaseindependent nuclease to induce single-stranded DNA nicks. J. Biol. Chem. 276:43285. 8. Fan, Z., P. J. Beresford, D. Y. Oh, D. Zhang, and J. Lieberman. 2003. Tumor suppressor NM23-H1 is a granzyme A-activated DNase during CTL-mediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell 112:659. 161 9. Fan, Z. S., P. J. Beresford, D. Zhang, Z. Xu, C. D. Novina, A. Yoshida, Y. Pommier, and J. Lieberman. 2003. Cleaving the oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nature Immunology 4:145. 10. Fan, Z. S., P. J. Beresford, D. Zhang, and J. Lieberman. 2002. HMG2 interacts with the nucleosome assembly protein SET and is a target of the cytotoxic T-lymphocyte protease granzyme A. Molecular and Cellular Biology 22:2810. 11. Han, J., L. A. Goldstein, B. R. Gastman, C. J. Froelich, X. M. Yin, and H. Rabinowich. 2004. Degradation of MCL-1 by granzyme B: Implications for bim-mediated mitochondrial apoptotic events. J. Biol. Chem. 12. Sebbagh, M., J. Hamelin, J. Bertoglio, E. Solary, and J. Breard. 2005. Direct cleavage of ROCK II by granzyme B induces target cell membrane blebbing in a caspaseindependent manner. J. Exp. Med. 201:465. 13. Trapani, J. A., and M. J. Smyth. 2002. Functional significance of the perforin/granzyme cell death pathway. Nat. Rev. Immunol. 2:735. 14. Ebnet, K., M. Hausmann, F. Lehmann-Grube, A. Mullbacher, M. Kopf, M. Lamers, and M. M. Simon. 1995. Granzyme A-deficient mice retain potent cell-mediated cytotoxicity. EMBO J. 14:4230. 15. Wilharm, E., J. Tschopp, and D. E. Jenne. 1999. Biological activities of granzyme K are conserved in the mouse and account for residual Z-Lys-SBzl activity in granzyme Adeficient mice. FEBS Lett. 459:139. 16. Heusel, J. W., R. L. Wesselschmidt, S. Shresta, J. H. Russell, and T. J. Ley. 1994. Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76:977. 17. Revell, P. A., W. J. Grossman, D. A. Thomas, X. Cao, R. Behl, J. A. Ratner, Z. H. Lu, and T. J. Ley. 2005. Granzyme B and the Downstream Granzymes C and/or F Are Important for Cytotoxic Lymphocyte Functions. J. Immunol. 174:2124. 18. Kam, C. M., D. Hudig, and J. C. Powers. 2000. Granzymes (lymphocyte serine proteases): characterization with natural and synthetic substrates and inhibitors. Biochim. Biophys. Acta 1477:307. 19. Hink-Schauer, C., E. Estebanez-Perpina, F. C. Kurschus, W. Bode, and D. E. Jenne. 2003. Crystal structure of the apoptosis-inducing human granzyme A dimer. Nat. Struct. Biol. 10:535. 20. Praveen, K., D. L. Evans, and L. Jaso-Friedmann. 2004. Evidence for the existence of granzyme-like serine proteases in teleost cytotoxic cells. J. Mol. Evol. 58:449. 21. Graves, S. S., D. L. Evans, and D. L. Dawe. 1985. Antiprotozoan activity of nonspecific cytotoxic cells (NCC) from the channel catfish (Ictalurus punctatus). J. Immunol. 134:78. 162 22. Jaso-Friedmann, L., D. S. Peterson, D. S. Gonzalez, and D. L. Evans. 2002. The antigen receptor (NCCRP-1) on catfish and zebrafish nonspecific cytotoxic cells belongs to a new gene family characterized by an F-box-associated domain. J. Mol. Evol. 54:386. 23. Jaso-Friedmann, L., and D. L. Evans. 1999. Mechanisms of cellular cytotoxic innate resistance in tilapia (Oreochromis nilotica). Dev. Comp Immunol. 23:27. 24. McKinney, E. C., and M. C. Schmale. 1994. Damselfish with neurofibromatosis exhibit cytotoxicity toward tumor targets. Dev. Comp Immunol. 18:305. 25. Suzumura, E., O. Kurata, N. Okamoto, and Y. Ikeda. 1994. Characteristics of Natural Killer-Like Cells in Carp. Fish Pathology 29:199. 26. Jaso-Friedmann, L., D. T. Harris, J. A. St, H. S. Koren, and D. L. Evans. 1990. A monoclonal antibody-purified soluble target cell antigen inhibits nonspecific cytotoxic cell activity. J. Immunol. 144:2413. 27. Faisal, M., I. I. Ahmed, G. Peters, and E. L. Cooper. 1989. Natural Cytotoxicity of Tilapia Leukocytes. Diseases of Aquatic Organisms 7:17. 28. Greenlee, A. R., R. A. Brown, and S. S. Ristow. 1991. Nonspecific cytotoxic cells of rainbow trout (Oncorhynchus mykiss) kill YAC-1 targets by both necrotic and apoptic mechanisms. Dev. Comp Immunol. 15:153. 29. Carlson, R. L., D. L. Evans, and S. S. Graves. 1985. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). V. Metabolic requirements of lysis. Dev. Comp Immunol. 9:271. 30. Evans, D. L., J. H. Leary, III, and L. Jaso-Friedmann. 1998. Nonspecific cytotoxic cell receptor protein-1: a novel (predicted) type III membrane receptor on the teleost equivalent of natural killer cells recognizes conventional antigen. Cell Immunol. 187:19. 31. Jaso-Friedmann, L., K. Praveen, J. H. Leary, III, and D. L. Evans. 2004. The gene and promoter structure of non-specific cytotoxic cell receptor protein-1 (NCCRP-1) in channel catfish (Ictalurus punctatus). Fish. Shellfish. Immunol. 16:553. 32. Heaton, M. P., N. L. LopezCorrales, T. P. L. Smith, S. M. Kappes, and C. W. Beattie. 1997. Directed cosmid isolation of bovine markers for physical assignment by fish. Animal Biotechnology 8:167. 33. Wingender, E., X. Chen, R. Hehl, H. Karas, I. Liebich, V. Matys, T. Meinhardt, M. Pruss, I. Reuter, and F. Schacherer. 2000. TRANSFAC: an integrated system for gene expression regulation. Nucleic Acids Res. 28:316. 34. Quandt, K., K. Frech, H. Karas, E. Wingender, and T. Werner. 1995. MatInd and MatInspector: new fast and versatile tools for detection of consensus matches in nucleotide sequence data. Nucleic Acids Res. 23:4878. 163 35. Kumar, S., K. Tamura, I. B. Jakobsen, and M. Nei. 2001. MEGA2: molecular evolutionary genetics analysis software. Bioinformatics. 17:1244. 36. Mount, S. M. 1982. A catalogue of splice junction sequences. Nucleic Acids Res. 10:459. 37. von Heijne G. 1986. A new method for predicting signal sequence cleavage sites. Nucleic Acids Res. 14:4683. 38. Wouters, M. A., K. Liu, P. Riek, and A. Husain. 2003. A despecialization step underlying evolution of a family of serine proteases. Mol. Cell 12:343. 39. Hink-Schauer, C., E. Estebanez-Perpina, E. Wilharm, P. Fuentes-Prior, W. Klinkert, W. Bode, and D. E. Jenne. 2002. The 2.2-A crystal structure of human pro-granzyme K reveals a rigid zymogen with unusual features. J. Biol. Chem. 277:50923. 40. Beresford, P. J., C. M. Kam, J. C. Powers, and J. Lieberman. 1997. Recombinant human granzyme A binds to two putative HLA-associated proteins and cleaves one of them. Proc. Natl. Acad. Sci. U. S. A 94:9285. 41. Huang, C., G. W. Wong, N. Ghildyal, M. F. Gurish, A. Sali, R. Matsumoto, W. T. Qiu, and R. L. Stevens. 1997. The tryptase, mouse mast cell protease 7, exhibits anticoagulant activity in vivo and in vitro due to its ability to degrade fibrinogen in the presence of the diverse array of protease inhibitors in plasma. J. Biol. Chem. 272:31885. 42. Huang, C., D. S. Friend, W. T. Qiu, G. W. Wong, G. Morales, J. Hunt, and R. L. Stevens. 1998. Induction of a selective and persistent extravasation of neutrophils into the peritoneal cavity by tryptase mouse mast cell protease 6. J. Immunol. 160:1910. 43. Xia, Z., C. M. Kam, C. Huang, J. C. Powers, R. J. Mandle, R. L. Stevens, and J. Lieberman. 1998. Expression and purification of enzymatically active recombinant granzyme B in a baculovirus system. Biochem. Biophys. Res. Commun. 243:384. 44. Kelly, J. M., M. D. O' Connor, M. D. Hulett, K. Y. Thia, and M. J. Smyth. 1996. Cloning and expression of the recombinant mouse natural killer cell granzyme Met-ase-1. Immunogenetics 44:340. 45. Irwin, D. M., K. A. Robertson, and R. T. MacGillivray. 1988. Structure and evolution of the bovine prothrombin gene. J. Mol. Biol. 200:31. 46. Pilat, D., T. Fink, B. Obermaier-Skrobanek, M. Zimmer, H. Wekerle, P. Lichter, and D. E. Jenne. 1994. The human Met-ase gene (GZMM): structure, sequence, and close physical linkage to the serine protease gene cluster on 19p13.3. Genomics 24:445. 47. Bell, G. I., C. Quinto, M. Quiroga, P. Valenzuela, C. S. Craik, and W. J. Rutter. 1984. Isolation and sequence of a rat chymotrypsin B gene. J. Biol. Chem. 259:14265. 164 48. Haddad, P., A. Wargnier, J. F. Bourge, M. Sasportes, and P. Paul. 1993. A promoter element of the human serine esterase granzyme B gene controls specific transcription in activated T cells. Eur. J. Immunol. 23:625. 49. Kamachi, Y., E. Ogawa, M. Asano, S. Ishida, Y. Murakami, M. Satake, Y. Ito, and K. Shigesada. 1990. Purification of a mouse nuclear factor that binds to both the A and B cores of the polyomavirus enhancer. J. Virol. 64:4808. 50. Wang, S. W., and N. A. Speck. 1992. Purification of core-binding factor, a protein that binds the conserved core site in murine leukemia virus enhancers. Mol. Cell Biol. 12:89. 51. Fregeau, C. J., and R. C. Bleackley. 1991. Transcription of two cytotoxic cell protease genes is under the control of different regulatory elements. Nucleic Acids Res. 19:5583. 52. Hanson, R. D., G. M. Sclar, O. Kanagawa, and T. J. Ley. 1991. The 5' -flanking region of the human CGL-1/granzyme B gene targets expression of a reporter gene to activated Tlymphocytes in transgenic mice. J. Biol. Chem. 266:24433. 53. Smyth, M. J., M. D. Hulett, K. Y. Thia, H. A. Young, T. J. Sayers, C. R. Carter, and J. A. Trapani. 1995. Cloning and characterization of a novel NK cell-specific serine protease gene and its functional 5' -flanking sequences. Immunogenetics 42:101. 54. Sattar, R., S. A. Ali, and A. Abbasi. 2003. Bioinformatics of granzymes: sequence comparison and structural studies on granzyme family by homology modeling. Biochem. Biophys. Res. Commun. 308:726. 55. Przetak, M. M., S. Yoast, and B. F. Schmidt. 1995. Cloning of cDNA for human granzyme 3. FEBS Lett. 364:268. 56. Bell, J. K., D. H. Goetz, S. Mahrus, J. L. Harris, R. J. Fletterick, and C. S. Craik. 2003. The oligomeric structure of human granzyme A is a determinant of its extended substrate specificity. Nat. Struct. Biol. 10:527. 57. Bode, W., Meyer E Jr, and J. C. Powers. 1989. Human leukocyte and porcine pancreatic elastase: X-ray crystal structures, mechanism, substrate specificity, and mechanism-based inhibitors. Biochemistry 28:1951. 58. Taylor, S. L., L. Jaso-Friedmann, A. B. Allison, A. Eldar, and D. L. Evans. 2001. Streptococcus iniae inhibition of apoptosis of nonspecific cytotoxic cells: a mechanism of activation of innate immunity in teleosts. Dis. Aquat. Organ 46:15. 59. Owen-Schaub, L. B., W. L. Crump, III, G. I. Morin, and E. A. Grimm. 1989. Regulation of lymphocyte tumor necrosis factor receptors by IL-2. J. Immunol. 143:2236. 60. Robinet, E., D. Branellec, A. M. Termijtelen, J. Y. Blay, F. Gay, and S. Chouaib. 1990. Evidence for tumor necrosis factor-alpha involvement in the optimal induction of class I allospecific cytotoxic T cells. J. Immunol. 144:4555. 165 61. Pham, C. T., D. A. Thomas, J. D. Mercer, and T. J. Ley. 1998. Production of fully active recombinant murine granzyme B in yeast. J. Biol. Chem. 273:1629. 62. Sun, J., C. H. Bird, M. S. Buzza, K. E. McKee, J. C. Whisstock, and P. I. Bird. 1999. Expression and purification of recombinant human granzyme B from Pichia pastoris. Biochem. Biophys. Res. Commun. 261:251. 63. Smyth, M. J., M. D. O' Connor, J. M. Kelly, P. Ganesvaran, K. Y. Thia, and J. A. Trapani. 1995. Expression of recombinant human Met-ase-1: a NK cell-specific granzyme. Biochem. Biophys. Res. Commun. 217:675. 64. Wilharm, E., M. A. Parry, R. Friebel, H. Tschesche, G. Matschiner, C. P. Sommerhoff, and D. E. Jenne. 1999. Generation of catalytically active granzyme K from Escherichia coli inclusion bodies and identification of efficient granzyme K inhibitors in human plasma. J. Biol. Chem. 274:27331. 65. Kummer, J. A., A. M. Kamp, F. Citarella, A. J. Horrevoets, and C. E. Hack. 1996. Expression of human recombinant granzyme A zymogen and its activation by the cysteine proteinase cathepsin C. J. Biol. Chem. 271:9281. 66. Sayers, T. J., A. R. Lloyd, D. W. McVicar, M. D. O' Connor, J. M. Kelly, C. R. Carter, T. A. Wiltrout, R. H. Wiltrout, and M. J. Smyth. 1996. Cloning and expression of a second human natural killer cell granule tryptase, HNK-Tryp-2/granzyme 3. J. Leukoc. Biol. 59:763. 67. Lockhart, B. E., J. R. Vencill, C. M. Felix, and D. A. Johnson. 2005. Recombinant human mast-cell chymase: an improved procedure for expression in Pichia pastoris and purification of the highly active enzyme. Biotechnol. Appl. Biochem. 41:89. 68. Griffiths, G. M., and S. Isaaz. 1993. Granzymes A and B are targeted to the lytic granules of lymphocytes by the mannose-6-phosphate receptor. J. Cell Biol. 120:885. 166 Figure 5.1. Genomic organization of TLGR-1 gene and promoter region. Exon sequences are represented in upper case letters, while introns and promoter region are represented as lower case letters. Putative transcription factor binding sites in the promoter region are represented as underlined bold letters with the name of the factor above the sequence. The predicted signal sequence at the N-terminus is highlighted while the dipeptide is represented with double underline. A putative glycosylation site is boxed and the polyadenylation signal is represented by underlined bold letters in the 3’ UTR. Triad residues constituting the active site are represented as bold letters in the sequence of amino acid chain. 167 1 gtctgtttttttaaacatccttcactgccaccttaaattcatttgatgtg 51 gctacatatttggtggtatgcatgaagagagaacacaagagagaaccgtc 101 aatagcaaaacctctacatgcccaccccctttgacagcagaagttcttat 151 ttccacagcttcacttatcagaagacaccaatcagattgtgatctgatta 201 tgatcatgctgagtcattttgtgtggttgtgtttgttcagggcttcattt AP-1 NFAT BRIGHT 251 taaatgctgctctcaccttttgtcatgtttccagttacacacttattaat 301 ttagcttttgtttcccatgttttgtccaaaaagcacacaaaagaaaaaag 351 caaaaggacattgccgattttttcttttttacattgctttaaaagacaaa 401 tgagcttattgaacatgtagatagttacactgattttaaaaaatccctat GATA-3 TATA-Box 451 aaacataaaaaatgtccaaatgtacatattaataacagactggttaagaa 501 cgtttgactgcaattcttttgtaattattttggtaaatgctactttcatg 551 catcatttattactgtattccacagtattccaacttccttatgtgttctt c-ETS-1 GATA-1 IRF-7 601 attatctgaaaattatttgcaaatacccgacttcattttcacatttcata IRF-2 651 aacaagcaaagtcttcgagccaacagccaataagatcactgcctcattcc 701 acttttcaacatgcagattacttactgaaataaataagcattgctgtttg 751 AP-1 tttatattgttgactgattcaatgacctttaagcgttttttcataacaga 801 agctgcaagagagcaaagagtcaaacctaaagcttgtatgtgtggttttt 851 cttactcatcatctactcatctagcattgggtttattgttgccgtgacag IRF-1 901 catcccacaaacaggctgaaggtttaagtttttcattttgagagcctgga NFAT 951 gaggcatttaaacaaaaaacactcgatagatgcaggaggaaaaaaatgtc STAT-1 1001 acaagcgcctcataacatgctatttccaggtagcacaaccacaggcgttg 1051 tatttactgaaacattcgtatggtctgtgatgcctcacagtttgactgac 1101 cgcacaggctcatcatagcgatccttttaaggttgcactcatgcagaaac TATA Box 1151 gatataagtaagaagtcaagggcaagcagatgaaaaccacagtctgaata 1201 1251 1301 1351 1401 M M H taaGGGTCTGACTCAACACCTCAAAGACGACAGGCTGAAAAGATGATGCA A V H D L M F V Y L L T C L G Q TGCTGTGCACGATCTTATGTTTGTCTATCTTCTAACATGTCTGGGACAAC H ATGgtaagattacaaaatgtaattacttatctaatgaaaacatccaacat G H G S E I I N taacttttttcctctcactcttgcagGACATGGAAGTGAAATCATAAATG G K N V P Q N S M Q Y M A S V Q I GCAAAAATGTCCCACAGAACTCAATGCAGTATATGGCTTCTGTGCAGATC 168 3 19 28 45 1451 1501 1551 1601 1651 1701 1751 1801 1851 1901 1951 2001 2051 2101 2151 2201 2251 2301 2351 2401 2451 2501 2551 D G K H V C G G F L V S E D F V L GATGGAAAACACGTATGTGGAGGATTTCTTGTCAGTGAAGACTTTGTGCT T A A H C Y K N CACGGCTGCACATTGTTACAAAAAgtaagtgaacaacctgtgtactttta tttgtttttctcaaaaaaacccagctaacacctacttaatattcgctcgc S P M E V V I G T H N L K K V tcagTTCTCCTATGGAGGTTGTAATTGGAACCCACAATCTGAAGAAGGTT N N N K M R Y S V K T C K H P R Y AATAACAACAAAATGAGATACAGTGTAAAGACATGCAAGCACCCACGATA D K V E S G N D I M L L K TGACAAAGTTGAGTCTGGTAATGACATCATGCTCCTCAAAgtaagtaaat atctttgaatgtgattcattacattacaataactccacttcgtacttcac caaagacttgatataagatcagaaatgtacagagttggtttctatcagtc L S R K L Q tttttattttttttccttttttggttcttctagCTCTCGAGGAAACTTCA L D K K V K P I Q L A R K E I K ACTGGACAAGAAAGTGAAACCGATACAACTGGCAAGGAAGGAGATTAAAG A K D N V K C Q V A G W G F T E T CAAAAGACAATGTAAAGTGTCAGGTGGCTGGATGGGGTTTCACAGAAACC N G K A V D V L R W V D V P L I D AATGGCAAAGCTGTTGATGTGCTGAGATGGGTAGATGTGCCTCTTATTGA L N V C K R K L K G K L P K G V CCTTAATGTCTGTAAGAGAAAATTGAAAGGCAAACTTCCAAAAGGTGTTA I C A G G S D T K N G F C Q TCTGTGCAGGAGGATCTGACACAAAGAATGGATTCTGCCAGgtatgtcct ctgttcttttaaagaaagtatcacttcacttgtagtattaaataaaagat G D S G G P L aaataaaacaagcttcttcccccttacagGGTGATTCTGGTGGCCCTCTG V C N G T A V G V V S F N I N G N GTGTGCAATGGAACAGCAGTTGGTGTTGTGTCTTTCAACATAAATGGAAA C K Y P N Y P N V Y T D I S K H TTGTAAATACCCAAATTACCCCAACGTCTATACAGATATATCAAAGCACC L S W I K N I L N K K Q C * TTTCCTGGATCAAAAACATTCTCAATAAAAAGCAATGCTAAGTATACACT GTAACAATGTGCAATCTGCAATATCTGGCATTATAATACTGTATGTGAAC TGCAGGATAAATGTAAAATAGCTTAAGACATCTCTCCTGTATTGCAAAGA TATTCCTGCTTGCTCTTAAATTAAAGTAAAAAATATTGAAGCTAAAAAAA AAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA 169 62 70 85 102 115 121 137 154 171 187 201 208 225 241 254 Figure 5.2. Predicted three dimensional model for TLGR-1. Four disulfide bonds which are conserved in other granzyme sequences are represented by circles. The extra Cys residue is marked with an arrow (One additional Cys residue at the C-terminus of the molecule is omitted from the model and not shown). 170 171 Figure 5.3. Comparison of TLGR-1 with other granzyme sequences. Multiple sequence alignments showing the conserved regions in TLGR-1 sequence, which represent the signature motifs for granzyme-like proteases. Conserved residues at the catalytic triad are represented with a black circle above the alignment. Darker shading represents identical or residues with similar properties in 100% of the sequences and lighter shading represents identical or residues with similar properties in 80% or more of all the sequences. 172 TLGR-1 CFGR1 AAQ54830 Human grnzA A31372 Human grnzB AAH30195 Human grnzK P49863 Human grnzM AAH25701 Human grnzH A32692 Mus grnzC NP_034501 Mus grnzD NP_034502 Mus grnzE NP_034503 Mus grnzF AAA37741 Mus grnzG NP_034505 : : : : : : : : : : : : IINGKNVPQNSMQYMASVQIDG----KHVCGGFLVSEDFVLTAAHCYKNSP-IIGGREVK-KPKPWMASVQSN--N--SHICGGTLIHQQWVLTAAHCKTFLQFK IIGGNEVTPHSRPYMVLLSLD----RKTICAGALIAKDWVLTAAHCNLNKR-IIGGHEAKPHSRPYMAYLMIWDQKSL-KRCGGFLIQDDFVLTAAHCWGSS--IIGGKEVSPHSRPFMASIQYG--G--HHVCGGVLIDPQWVLTAAHCQYRFTKG IIGGREVIPHSRPYMASLQRNG----SHLCGGVLVHPKWVLTAAHCLAQR--M IIGGHEAKPHSRPYMAFVQFLQEKSR-KRCGGILVRKDFVLTAAHCQGSS--IIGGNEISPHSRPYMAYYEFLKVGGKKMFCGGFLVRDKFVLTAAHCKGRS--IIGGHVVKPHSRPYMAFVMSVDIKGNRIYCGGFLIQDDFVLTAAHCKNSS--IIGGHVVKPHSRPYMAFVKSVDIEGNRRYCGGFLVQDDFVLTAAHCRNRT--IIGGHEVKPHSRPYMARVRFVKDNGKRHSCGGFLVQDYFVLTAAHCTGSS--IIGGHEVKPHSRPYMAFIKSVDIEGKKKYCGGFLVQDDFVLTAAHCRNRS--- : : : : : : : : : : : : 47 48 47 49 49 47 49 50 50 50 50 50 TLGR-1 CFGR1 AAQ54830 Human grnzA A31372 Human grnzB AAH30195 Human grnzK P49863 Human grnzM AAH25701 Human grnzH A32692 Mus grnzC NP_034501 Mus grnzD NP_034502 Mus grnzE NP_034503 Mus grnzF AAA37741 Mus grnzG NP_034505 : : : : : : : : : : : : --MEVVIGTHNLK-KVNNNKMRYSVKTCKHPRYDKV-ESGNDIMLLKLSRKLQ P-IEVLLGAHSLT-KDKNAMRVKVLCFHISPKFSAT-TRVHDIMLLKLQDKVQ --SQVILGAHSITREEPTKQIMLVKKEFPYPCYDPA-TREGDLKLLQLTEKAK --INVTLGAHNIKEQEPTQQFIPVKRPIPHPAYNPK-NFSNDIMLLQLERKAK QSPTVVLGAHSLSKNEASKQTLEIKKFIPFSRVTSD-PQSNDIMLVKLQTAAK AQLRLVLGLHTLD---SPGLTFHIKAAIQHPRYKPVPALENDLALLQLDGKVK --INVTLGAHNIKEQERTQQFIPVKRPIPHPAYNPK-NFSNDIMLLQLERKAK --MTVTLGAHNIKAKEETQQIIPVAKAIPHPDYNPD-DRSNDIMLLKLVRNAK --MTVTLGAHNITAKEETQQIIPVAKDIPHPDYNAT-IFYSDIMLLKLESKAK --MTVTLGAHNIKAKEETQQIIPVAKAIPHPDYNAT-AFFSDIMLLKLESKAK --MRVILGAHNIRAKEETQQIIPVAKAIPHPAYDDK-DNTSDIMLLKLESKAK --MTVTLGAHNIKAKEETQQIIPVAKAIPHPAFNRK-HGTNDIMLLKLESKAK : : : : : : : : : : : : 96 98 97 99 101 97 99 100 100 100 100 100 TLGR-1 CFGR1 AAQ54830 Human grnzA A31372 Human grnzB AAH30195 Human grnzK P49863 Human grnzM AAH25701 Human grnzH A32692 Mus grnzC NP_034501 Mus grnzD NP_034502 Mus grnzE NP_034503 Mus grnzF AAA37741 Mus grnzG NP_034505 : : : : : : : : : : : : LDK-KVKPIQLARKEIKAKDNVKCQVAGWGFTETN-GKAVDVLRWVDVPLIDL LKKNKVDVKKIPKSGKDIPAGTKCEVRGWGTTHVKNPKACDTLQELEVTVVDR INK-YVTILHLPKKGDDVKPGTMCQVAGWGRTHN-SASWSDTLREVNITIIDR RTR-AVQPLRLPSNKAQVKPGQTCSVAGWGQTAP-LGKHSHTLQEVKMTVQED LNK-HVKMLHIR-SKTSLRSGTKCKVTGWGATDPDSLRPSDTLREVTVTVLSR PSR-TIRPLALPSKRQVVAAGTRCSMAGWGLTHQ-GGRLSRVLRELDLQVLDT WTT-AVRPLRLPSSKAQVKPGQLCSVAGWGYVS--MSTLATTLQEVLLTVQKD RTR-AVRPLNLPRRNAHVKPGDECYVAGWGKVTP-DGEFPKTLHEVKLTVQKD RTK-AVRPLKLPRSNARVKPGDVCSVAGWGSRSINDTKASARLREVQLVIQED RTK-AVRPLKLPRPNARVKPGDVCSVAGWGPRSINDTKASARLREAQLVIQED RTK-AVRPLKLPRPNARVKPGHVCSVAGWGRTSINATQRSSCLREAQLIIQKD RTK-AVRPLKLPRPNARVKPGDVCSVAGWGKTSINATKASARLREAQLIIQED : : : : : : : : : : : : 147 151 148 150 152 148 149 151 152 152 152 152 TLGR-1 CFGR1 AAQ54830 Human grnzA A31372 Human grnzB AAH30195 Human grnzK P49863 Human grnzM AAH25701 Human grnzH A32692 Mus grnzC NP_034501 Mus grnzD NP_034502 Mus grnzE NP_034503 Mus grnzF AAA37741 Mus grnzG NP_034505 : : : : : : : : : : : : NVCKRKLKG----KLPKGVICAGGSDTKNGFCQGDSGGPLVCNG--TAVGVVS ELCNCYYNSK--PTITANMLCAGNKQRDKDACWGDSGGPLECKK--NIVGVVS KVCNDRNHYNFNPVIGMNMVCAGSLRGGRDSCNGDSGSPLLCEG--VFRGVTS RKCESDLRH---YYDSTIELCVGDPEIKKTSFKGDSGGPLVCNK--VAQGIVS KLCNSQSYYNGDPFITKDMVCAGDAKGQKDSCKGDSGGPLICKG--VFHAIVS RMCNNSRFWN--GSLSPSMVCLAADSKDQAPCKGDSGGPLVCGKGRVLAGVLS CQCERLFHG---NYSRATEICVGDPKKTQTGFKGDSGGPLVCKD--VAQGILS QVCESQFQS---SYNRANEICVGDSKIKGASFEEDSGGPLVCKR--AAAGIVS EECKKRFR----YYTETTEICAGDLKKIKTPFKGDSGGPLVCHN--QAYGLFA EECKKRFR----HYTETTEICAGDLKKIKTPFKGDSGGPLVCDN--KAYGLLA KECKKYFY----KYFKTMQICAGDPKKIQSTYSGDSGGPLVCNN--KAYGVLT EECKKLWY----TYSKTTQICAGDPKKVQAPYEGESGGPLVCDN--LAYGVVS : : : : : : : : : : : : 194 200 199 198 203 199 197 199 199 199 199 199 TLGR-1 CFGR1 AAQ54830 Human grnzA A31372 Human grnzB AAH30195 Human grnzK P49863 Human grnzM AAH25701 Human grnzH A32692 Mus grnzC NP_034501 Mus grnzD NP_034502 Mus grnzE NP_034503 Mus grnzF AAA37741 Mus grnzG NP_034505 : : : : : : : : : : : : FNINGNCKYPNYPN-VYTDISKHLSWIKNILNKKQCGGSGCGNPKK--PGVYTLLSKEHIDWINKIIKK---FGLENKCGDPRGPGVYILLSKKHLNWIIMTIKGAV-YGRNNGMP----PR-ACTKVSSFVHWIKKTMKRH--GGHECGVATK--PGIYTLLTKKYQTWIKSNLVPPHTN FSSRVCTDIFKPP--VATAVAPYVSWIRKVTGRSA-YGNKKGTPPG-----VYIKVSHFLPWIKRTMKRL--YGQTDGSAPQ-----VFTRVLSFVSWIKKTMKHS--YAKNGTISSG-----IFTKVVHFLPWISWNMKLL--YAKNRTISSG-----VFTKIVHFLPWISRNMKLL--YGLNRTIGPG-----VFTKVVHYLPWISRNMKLL--YGINRTITPG-----VFTKVVHFLPWISTNMKLL--- 173 : : : : : : : : : : : : 229 231 234 227 238 232 226 228 228 228 228 228 Figure 5.4. Phylogenetic analysis of TLGR-1. Phylogram represents the evolutionary position of TLGR-1 compared to related serine proteases. The tree was derived by parsimony analysis, with Mega version 2.1. Sequences of mature protease were used in the analysis and accession numbers are provided after the name. Numbers shown above the branches are bootstrap values based upon 1000 replicates for parsimony. The tree was rooted on a sub-tree containing complement factor D to determine the clustering of TLGR-1 with similar granzyme. 174 98 Mus grnzB NP 038570 Rat grnzB NP 612526 Mus grnzC NP 034501 Human grnzB AAH30195 98 Human grnzH A32692 Mus grnzF AAA37741 83 Mus grnzG NP 034505 99 Mus grnzD NP 034502 90 87 Mus grnzE NP 034503 Cow granzB NP 776721 Mus cathepsin G CAA55290 99 100 Human cathepsin G AAA51919 Rat grnzJ AAC53168 100 100 Mus mast cell chymase AAA39492 Rat mast cell chymase NP 037224 Human mast cell chymase NP 001827 Mus mast cell chymase 2 P15119 97 Mus mast cell chymase 10 AAK51074 88 Mus mast cell chymase 4 NP 034909 76 100 99 Rat mast cell chymase 1 P09650 CFGR2 EST:CB940297 Danio hypothtical CFGR4 EST:CB940725 TLGR-1 96 100 CFGR3 EST: CB940628 Pig Cap37 P80015 Human Cap37 P20160 Human myeloblasin elastase P24158 100 Mus myeloblasin elastase Q61096 99 Mus neut elastase NP 056594 100 Human neut elastase AAA36359 100 Mus grnzK AAC17930 100 Rat grnzK NP 058815 Human grnzK P49863 CFGR1 AAQ54830 Fugu hypothetical Xenopus GrnzA CAD66429 Chick grnzA CAD66428 Human grnzA A31372 100 Mus grnzA NP 034500 Human grnzM AAH25701 Rat grnzM Q03238 100 100 Mus grnzM NP 032530 100 Rat comp factor D I55608 Mus comp factor D WMMS28 Pig comp factor D P51779 100 99 175 Human comp factor D AAH57807 Figure 5.5. Analysis of tissue expression of TLGR-1. RT-PCR analysis for TLGR-1 using cDNA from various tilapia tissues. Lane 1, purified NCC from peripheral blood; lane 2, total peripheral blood leukocytes; lane 3, total blood; lane 4, muscle; lane 5, liver; lane 6, gill; lane 7, spleen; lane 8, kidney; lane 9, TMB8 (a tilapia continuous cell line TMB-8 was provided by Dr R. Hedrick, University of California-Davis. These cells were originally established from Oreochromis mossambicus cardiac tissue). Beta actin was used as a normalizer. 176 177 Figure 5.6. Transcriptional regulation of TLGR-1 in NCC. Real-time RT-PCR analysis of cDNA from NCC purified from tilapia peripheral blood after subjecting to different treatments. (a) NCC were treated with recombinant tilapia TNF-alpha and incubated for two or four hours and analyzed for TLGR-1 expression. (b) NCC treated with PMA and calcium ionophore (A23187) for two or four hours. (c) NCC treated with lipopolysaccharides (from E. coli). 178 A 30 Fold Increase 25 20 15 10 5 0 0H 2H 4H Time B 2.5 Fold Increase 2 1.5 1 0.5 0 0H 2H 4H Time C 7 Fold Increase 6 5 4 3 2 1 0 0H 2H Time 179 4H Figure 5.7. Production of recombinant TLGR-1 in Pichia pastoris. (a) Representation of expression cassettes engineered to generate pro and mature TLGR-1 using pPICZ-alpha expression vector. Pro and mature forms of TLGR-1 were expressed as fusion proteins starting with the yeast alpha factor at the N-terminus and poly-histidine and c-myc tagged granzyme at the C-terminus. Presence of a Kex2 site in between the alpha factor and the granzyme allowed the expression of granzyme with a native N-terminus, which can be secreted into the culture medium with minimum contaminating yeast proteins. (b) Optimization of recombinant protein expression. The culture supernatants were collected at the time points indicated and subjected to TCA precipitation followed by Western blotting. His-tagged proteins were detected with INDIAHis probe conjugated to HRP followed by chemiluminescence. Yeast trsansfected with empty vector alone (E) also were checked for the expression of proteins and only 72 hour time point is shown. 180 A B 181 Figure 5.8. Purification of pro and mature TLGR-1 recombinant proteins. Supernatants were collected after methanol induction for 72 hours and total proteins were concentrated by ammonium sulfate precipitation. His-tagged proteins were purified by nickel affinity chromatography. Purification success was verified by Western blotting and detection with INDIA-His probe. Lane V: vector alone, lane F: flow though from the column, lane E1, E2, and E3: three consecutive elutions of the column using buffer containing 250 mM imidazole. 182 183 Figure 5.9. Enzymatic activity of recombinant TLGR-1. (a) Plot depicting the change in optical density against time from an in vitro enzyme kinetic assay using N-succinyl-Phe-Leu-Phe-SBzl as a substrate. At a final concentration of 0.3 mM. Enzyme concentration was approximately 1 nM for both mature and pro-TLGR-1. (b) Hydrolysis of N-succinyl-Ala-Ala-Pro-Phe-pNA by mature and pro-TLGR-1. 184 A B 185 Figure 5.10. Substrate specificity of TLGR-1. The enzymatic activity of mature and pro-TLGR-1 were measured using the indicated thiobenzyl ester substrates in final concentrations of 0.3 mM. Enzyme concentration was approximately 1 nM for both mature and pro-TLGR-1. Rate of hydrolysis is depicted as change in mOD per minute. 186 Boc-Ala-Ala-Asp-SBzl Boc-Ala-Ala-Met-SBzl N-Succinyl-Phe-LeuPhe-SBzl Empty Vector Pro-TLGR TLGR Z-Arg-SBzl Z-Lys-SBzl 0 2 4 6 8 Change in mOD/min 187 10 12 Table 5.1: Organization of TLGR-1 gene. Amino acid numbering based on the mature polypeptide chain. Segment Size Amino acids Features Exon 1 100 bp -25 to -5 5’ untranslated region, Signal peptide Exon 2 148 bp -4 to 28 Activation dipeptide (Ser and Glu) and N-terminus of mature protein, Active site His residue Exon 3 136 bp 29 to 73 Active site Asp residue Exon 4 258 bp 74 to 159 First of the substrate specificity pocket triplet residues (Gly) Exon 5 196 bp (up to polyadenylation signal) 160 to 212 Active site Ser residue, second and third of the substrate specificity pocket triplet residues (Asn and Asn), 3’ untranslated region, polyadenylation signal 188 Table 5.2: Pairwise comparison of TLGR-1 with related serine proteases. Protein CFGR-1 Human Granzyme A Mus Granzyme A Human Granzyme B Mus Granzyme B Rat Granzyme B Mus Granzyme C Mus Granzyme D Mus Granzyme E Mus Granzyme F Mus Granzyme G Human Granzyme H Rat Granzyme J Human Granzyme K Mus Granzyme K Rat Granzyme K Human Granzyme M Mus Granzyme M Rat Granzyme M Mus mast cell chymase Human Cathepsin G Mus Cathepsin G Human Complement factor D Accession # AAQ54830 A31372 NP_034500 AAH30195 NP_038570 NP_612526 NP_034501 NP_034502 NP_034503 AAA37741 NP_034505 A32692 AAC53168 P49863 AAC17930 NP_058815 AAH25701 NP_032530 Q03238 P15119 AAA51919 CAA55290 AAH57807 189 % Identity % Similarity 37.1 34.5 31.6 34.6 38.0 39.1 34.9 35.3 35.7 36.2 38.3 33.0 35.0 33.7 36.2 37.0 33.2 32.4 32.6 35.5 34.9 32.8 31.8 47.7 47.2 45.7 44.4 48.3 48.5 46.4 46.4 46.4 46.4 48.9 41.6 45.3 46.5 47.7 46.6 50.2 47.5 47.3 45.7 46.9 43.3 46.4 CHAPTER 6 CONSTITUTIVE EXPRESSION OF TUMOR NECROSIS FACTOR-ALPHA IN CYTOTOXIC CELLS OF TELEOSTS AND ITS ROLE IN REGULATION OF CELLMEDIATED CYTOTOXICITY1 1 Reprinted from publication Praveen, K., D. L. Evans and L. Jaso-Friedmann. 2005. Constitutive expression of tumor necrosis factor-alpha in cytotoxic cells of teleosts and its role in regulation of cell-mediated cytotoxicity. Molecular Immunology, in press. ©2005, Reprinted here with permission from Elsevier. 190 ABSTRACT Cytotoxic T lymphocytes (CTL) and natural killer (NK) cells are the main killer cell populations of the immune system. The mechanisms by which these cells recognize target cells vary considerably, while the effector molecules used to facilitate target cell death are highly conserved. The main pathways utilized by killer cells consist of granule exocytosis and those mediated by members of the TNF superfamily. Nonspecific cytotoxic cells (NCC) are the first identified cytotoxic cell population in teleosts. We have previously demonstrated the expression of granzymes and Fas ligand in these cells. This is the first report of the expression of tumor necrosis factor-alpha in these killer cells. A cDNA coding for TNF was cloned and sequenced from NCC purified from Nile tilapia (Oreochromis niloticus). Factors regulating the transcriptional modulation of TNF in these cells were identified by RT-PCR analysis. The mature form of tilapia TNF was expressed as a recombinant protein and biological activities were analyzed. Using a cross-reacting anti-TNF polyclonal antibody, analysis of TNF expression suggested that tilapia NCC constitutively express the membrane-bound as well as secreted forms of TNF. Recombinant tilapia TNF effectively induced cytotoxicity in the mammalian cell line WEHI, although to a lesser extent compared to the murine TNF. Treatment with recombinant TNF protected NCC from activation-induced cell death. Recombinant tilapia TNF was also effective in upregulation of granzyme transcription in tilapia NCC. These data suggest that teleost TNF may play a role in diverse effector functions of cytotoxic cells from ectotherms, similar to the biological functions described for mammalian TNF. Keywords: Tumor Necrosis Factor-alpha, Cytotoxicity, Nonspecific Cytotoxic Cells, Activation-induced cell death, Stress Activated Serum Factors 191 1. INTRODUCTION Apoptotic cellular death is initiated by two main signaling mechanisms called the intrinsic and extrinsic pathways. The intrinsic pathway triggers apoptosis as a result of DNA damage, cell cycle checkpoint defects, loss of survival factors and other types of severe cellular stress conditions. In all of those circumstances death occurs as a consequence of the activation of the pro-apoptotic arm of the Bcl-2 gene superfamily. This engages mitochondria to cause the cytosolic release of apoptogenic factors such as cytochrome C and SMAC/DIABLO (Adams and Cory, 1998, Green, 2000, Hunt and Evan, 2001). The extrinsic pathways of cellular death are initiated by binding of members of the TNF superfamily ligands to their death receptors (Ashkenazi, 2002). The TNF ligand super-family is composed of members such as TNF, Fas ligand and TRAIL that are grouped by their structural and functional similarities. These ligands recognize different specific receptors, which together constitute the TNF receptor super family (Kwon et al., 1999, Locksley et al., 2001, Wallach et al., 1999). TNF binds to two membrane receptors of 55-60 kDa (TNF-R1) and 75-80 kDa (TNF-R2). TNF-R1 is widely expressed on a variety of cell types and mediates most of the TNF functions. TNF-R2 expression is tightly regulated and restricted primarily to cells of lymphoid tissue (Grell et al., 1995b). Like other members of the TNF super-family, TNF is synthesized as type II transmembrane protein that may be expressed in either of two forms: the membrane embedded “pro” molecule or the cleaved “mature” form (Idriss and Naismith, 2000a). Both forms are active and self assemble into non-covalent trimers, whose individual chains fold into compact jelly roll β-sandwiches and interact at hydrophobic interfaces (Fesik, 2000). 192 The physiological significance and difference between the membrane and soluble forms of TNF super-family members has been the focus of many investigations. Recombinant soluble TNF-α and lymphotoxin are capable of inducing apoptotic cell death in mouse as well as human T lymphocyte blasts in vitro (Sarin et al., 1995b). It has also been demonstrated that the membrane-bound TNF and FasL are more potent inducers of cytotoxicity than their soluble trimeric forms (Eissner et al., 1995, Grell et al., 1995a, Schneider et al., 1998, Tanaka et al., 1998). Membrane-bound FasL was cytotoxic for human peripheral blood lymphocytes, while the soluble form blocked the killing (Suda et al., 1997). Although these studies have shed light into the possible mechanisms of actions of the different forms of TNF, the role of TNF alpha in immune functions is still unclear. In mammalian NK cells, expression of TNF alpha alone was not sufficient to induce tumor cell killing while simultaneous interactions of at least three TNF family ligands (TNF, LT- 1 2 and FasL) with their corresponding receptors effectively generated pro-apoptotic signals in tumor cells (Kashii et al., 1999). Teleost TNFs have been sequenced and expressed from a variety of species (GarciaCastillo et al., 2002, Hirono et al., 2000, Laing et al., 2001, Saeij et al., 2003, Zou et al., 2003b). Comparisons of the gene structure and phylogenetic analysis of the amino acid sequences appear to indicate that the teleost TNFs are more closely related to mammalian TNF alpha than beta (Goetz et al., 2004). It has been suggested that fish do not have a TNF beta lymphotoxin-like gene. Although this could suggest a more prevalent role for TNF alpha in fish species, there are many unanswered questions about its functional role. While it is clear that fish TNF is produced by macrophages in response to bacterial stimulus, other cell sources for this cytokine have not been determined (Goetz et al., 2004). Most of the studies on teleost TNF have utilized mixed populations of myeloid cells, which make the functional characterization difficult to achieve 193 (Goetz et al., 2004). Using long-term hematopoietic cell-lines, expression of TNF in channel catfish was shown to be mostly in T cells and macrophages, but not in B cells and fibroblasts (Zou et al., 2003b). In the present study we have cloned and expressed the TNF-alpha gene from freshly purified nonspecific cytotoxic cells (NCC) of tilapia. NCC have been well characterized in a number of fish species and they appear to be functionally related to mammalian NK cells (Carlson et al., 1985, Evans et al., 1984, Faisal et al., 1989, Graves et al., 1985, Greenlee et al., 1991, Jaso-Friedmann et al., 2002, Jaso-Friedmann and Evans, 1999, McKinney and Schmale, 1994). We have previously shown that tilapia NCC expressed a molecule functionally characterized as Fas ligand and we have molecular information about teleost granzymes. (Bishop et al., 2002, Jaso-Friedmann et al., 2000a, Praveen et al., 2004). However, this is the first report on the molecular identity of a death ligand in tilapia. Our results show that, much like its mammalian counterpart, TNF alpha from tilapia NCC appears to have pleiotropic immunological functions. 2. MATERIALS AND METHODS 2.1. Experimental animals and isolation of NCC. Outbred tilapia (Oreochromis niloticus) weighing 60-100 g were obtained from Americulture, Inc. Animas, NM. Fish were maintained in fiberglass aquaria equipped with a constant flow through system at 23-25o C. Fish were fed a commercial diet of pelleted fish food (Southern States Co-operative Inc, VA). Fish were acclimatized for a minimum of 3 months prior to the study and were free from any clinical infections. NCC were purified from peripheral blood, anterior kidney or spleen of tilapia as previously described (Bishop et al., 2000). Purity of cell preparation was verified by flowcytometric analysis using 5C6 (a monoclonal antibody 194 detecting NCCRP-1, which is an activation marker found exclusively on NCC (Evans et al., 1998, Jaso-Friedmann et al., 2002). 2.2. Construction and screening of tilapia cDNA library Construction of cDNA libraries from NCC of different fish species has been previously described (Praveen et al., 2004). Conserved regions in various teleost TNF-alpha sequences were identified by multiple sequence alignments using Clustal W provided with vector NTI package, version 6 (InforMax Inc). Two degenerate primers (TNFDGN1F: 5’GNGCHAARGCHGCHATYCA-3’ and TNFDGN3R: 5’-CARRTARATDGCRTTRTACCA-3’) were used to amplify a portion of tilapia TNF, using tilapia NCC cDNA library as template. Amplicons were TA cloned in to pDrive cloning vector using a PCR cloning kit (Qiagen, Valencia, CA). Inserts were sequenced in two directions and compared with the known sequences in DDBJ/EMBL/GenBank databases using BLAST version 2.2.5 (Altschul et al., 1997). Sequence from one of the amplicons which had closest similarity to the known TNF sequences was used to design primers to amplify the entire 5' and 3' ends of the tilapia TNF as described previously (Praveen et al., 2004). 2.3. Phylogenetic analysis and protein modeling Similar analyses as those previously done with catfish granzyme and NCCRP-1 of zebrafish were performed (Jaso-Friedmann et al., 2002, Praveen et al., 2004). The three dimensional structure of tilapia TNF was modeled using SWISS-MODEL in the first approach mode accessible via the internet (http://www.expasy.org/swissmod). The co-ordinate files were 195 imported to RasWin software version 2.6 for analyzing bond lengths and other conformational features of the molecule. 2.4. RT-PCR analysis Purified NCC were subjected to various treatments and total RNA was isolated using RNeasy Mini Kit (Qiagen, Valencia, CA) according to manufacturer’s recommendations. Synthesis of cDNA was done using a cDNA synthesis kit (Invitrogen, Carlsbad, CA) using olido-dT primer. Primers used for the PCR amplification were as follows: TLTNFRTF1: 5’ggttagttgagaagaaatcacctgca-3’ and TNTNFRTR1: 5’-gtcgtcgctattcccgcagatca-3’. PCR was done with initial 2 minutes denaturation at 94oC followed by 25 cycles of following cycle: denaturation at 94oC for 30 s, annealing at 57oC for 30 s, extension at 72oC for 30 s. The products were resolved on a 1.5% agarose gel and ethidium bromide-stained bands were quantified using a Kodak DC290 digital camera and digitized using UN-SCAN-IT software (Silk Scientific, Orem, UT). Band intensities (pixel values) were normalized to that of beta-actin. 2.5. Cloning and expression of recombinant tilapia TNF A region at the C-terminus, which corresponds to the mature extracellular domain of tilapia TNF was amplified by PCR using a proofreading DNA polymerase (Ex-Taq: Takara Mirus Bio, Madison, WI). The forward PCR primer was designed to introduce a 6-His-Tag and a Nhe I site at the 5’ end (TNFRECF1: 5’CGTAGCTAGCCATCACCATCACCATCACCTAAAACGGATCAGCAGCA-3’), while the reverse primer was engineered to bear an Xho I site (TNFRECR1: 5’CTAGCTCGAGTCAAAGTGCAAACACGCCAAAGAAGGTC-3’). The stop codon of tilapia 196 TNF was kept intact to ensure a native C-terminus for the recombinant protein. A full-length tilapia TNF cDNA clone, whose sequence was verified by sequencing both the strands, was used as a template for PCR reaction. The amplicon was purified and directionally cloned in to the pET-21b(+) expression vector (Novagen, San Diego, CA) between Nhe I and Xho I sites. The recombinant plasmid (pET-21-TLTNF) was sequenced in both direction and was electroporated in to expression strain of E. coli (BL21-DE3: Novagen, San Diego, CA). Bacteria transformed with pET-21-TLTNF and empty vector were cultured in LB-ampicillin medium to an OD600 of 0.6-1.0. Production of recombinant protein was induced with 1 mM isopropyl-beta-Dthiogalactopyranoside (IPTG: Fisher Scientific, Fair Lawn, NJ). Optimization of protein expression was verified by harvesting bacteria at various time points by centrifugation of culture medium followed by lysis of the pellet and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Lysates were prepared from IPTG-induced cultures by sequential incubations in lysozyme (1 mg/ml), Triton X-100 (0.5%), DNAse I (5 µg/ml) and RNAse A (10 µg/ml) in extraction buffer containing protease inhibitors (Sigma, St. Louis, MO). Recombinant His6-tagged tilapia TNF was purified by metal affinity chromatography using Ni-NTA agarose (Qiagen, Valencia, CA) according to manufacturer’s instructions. The positive fractions were pooled, desalted and concentrated using Centricon YM-3 centrifugal filters (Millipore, Billerica, MA). Aliquots were stored at -80oC. Protein concentrations were estimated using Biorad Protein Assay Kit (Biorad, Hercules, CA). 2.6. Identification of antibody binding to tilapia TNF Recombinant tilapia TNF was boiled in sample buffer and was resolved on a 12% acrylamide gel and transferred on to nitrocellulose membrane. Membranes were probed with 197 commercially available TNF antibodies and control antibodies (anti-Fas ligand). A polyclonal antibody raised against mouse TNF-alpha (P350: Pierce, Rockford, IL) was identified as the only cross-reacting antibody. 2.7. Flowcytometry Tilapia NCC from various tissues were purified as previously described. Constitutive expression of TNF in NCC was analyzed by one and two-color flowcytometry using an EPICS XL-MCL analyzer (Coulter Electronics). NCC were incubated in PBS containing BSA and 0.05% Sodium Azide (PAB) before staining with polyclonal anti-TNF antibody and FITC conjugated anti-rabbit IgG (Sigma Immunochemicals). Control rabbit IgG and appropriate conjugate controls were used to differentiate the non-specific binding of mAbs in the permeabilized cells. For the two color analysis, NCCRP-1 expression was detected by staining cells with mAb 5C6 (Evans et al., 1998, Jaso-Friedmann and Evans, 1999) followed by biotinylated anti-Mouse IgM (Sigma Immunochemicals) and phycoerythrin conjugated streptavidin. 2.8. Cytotoxicity Ability of recombinant tilapia TNF to kill TNF-sensitive targets was assessed using a murine fibrosarcoma cell line WEHI-164S (ATCC, Manassas, VA). The cells were seeded into flat bottom 96-well microplate (5 x 104/well) and grown to confluent monolayers in RPMI-1640 supplemented with L-glutamine, sodium pyruvate, MEM vitamin solution, MEM amino acid solution, MEM nonessential solution (Cellgro Media Tech, Washington, DC), 50 mg/ml gentamicin and 10% fetal bovine serum (FBS). After overnight culture at 37oC under 5% CO2, 198 the medium was removed and replaced with 100 µl RPMI-1640 containing 10% FBS and 8 µg/ml of actinomycin D (Sigma, St. Louis, MO). Serially diluted recombinant tilapia TNF (100 µl volume) in RPMI was added to each well and incubated for 16 hours. The induction of cytotoxicity was assessed by DNA hypoploidy analysis. For chromium release assay, WEHI cells (2×106 cells/ml) were labeled with 20 µCi of sodium chromate (Amersham, IL) for 3 h at 37°C. Cells were washed three times in medium containing 8 µg/ml of actinomycin D and diluted to 1×105 cells/ml. Labeled targets were mixed with effector cells (tilapia NCC) at different E:T ratios and the plates were centrifuged at 500 x g. In order to demonstrate the specificity of killing, NCC were incubated with anti-TNF polyclonal antibody or control rabbit IgGs for 2 hours, before mixing with the target cells. After 18 hours of incubation 100 µl of each supernatant was removed to determine radioactivity (Beckman Biogamma II counter; Beckman Instruments, CA). The results are expressed as percentage specific release (SR). Percent SR = (Test Release – Spontaneous Release/ Total Release – Spontaneous release) ×100. 2.9. DNA hypoploidy analysis Fragmentation of DNA in WEHI cells, induced by recombinant TNF was determined as described before for Tetrahymena (Jaso-Friedmann et al., 2000a). Briefly, treatments were stopped by the addition of nuclear isolation media (NIM, propidium iodide [50 µg/ml], Triton X100 [0.01%] and RNase [1 mg/ml] in phosphate-buffered saline, pH 7.3). Cells were resuspended in NIM for 10 min at 4oC in the dark. The isolated nuclei were then analyzed for DNA content using flowcytometry. 199 3. RESULTS 3.1. Molecular cloning of TNF cDNA from tilapia NCC Highly conserved regions in the known TNF sequences were used to design degenerate primers to amplify a portion of tilapia TNF. The PCR reactions using cDNA library and fresh cDNA from tilapia NCC as a template yielded a band of expected size. The PCR product was purified and cloned into pDrive TA cloning vector and sequenced to verify the identity. This product had highest similarity to known TNF-alpha sequences. Using this sequence information, non-degenerate primers were designed to obtain full-length tilapia TNF cDNA by 5’ and 3’ RACE as described before (Praveen et al., 2004). The complete coding sequence of tilapia TNF is shown in Fig. 1 (NCBI accession # AY428948). The cDNA contains an open reading frame of 744 bp encoding a polypeptide of 247 residues with a predicted molecular weight of 27488.28 Da. The length of the 5’ untranslated region (UTR) is 114 bp, while 3’ UTR has a length of 448 bp. Tilapia TNF cDNA has a polyadenylation signal (AATAAA) and multiple mRNA instability motifs in the 3’ UTR. The signal peptide prediction algorithms using McGeoch's (McGeoch, 1985) or von Heijne's (von Heijne G., 1986) methods failed to detect any signal peptide in tilapia TNF. However, using Singer's classification for membrane topology (Singer, 1990), a transmembrane domain (residues numbers 35-51, Fig. 1) can be predicted with N-terminus inside the cytoplasm, giving an indication for the production of tilapia TNF as a membrane-bound form. Comparison of known TNF sequences revealed that many of the signature residues are conserved in tilapia TNF (Fig. 2a). Most of the sequence identities are confined to the C-terminal domain, as reported for other fish sequences. The site used by mammalian TNF-alpha converting enzyme to release the mature (secreted) form of TNF is conserved in all the fish sequences. The 200 two cysteine residues, which are crucial in the correct folding of the mature TNF is found to be conserved in the majority of sequences (Fig. 2a). Thus, the overall predicted structure of mature tilapia TNF was compared with other known 3D structures of mammalian TNF (Fig. 2b). Using this model, we could predict similar biological functions of tilapia TNF like its mammalian counterparts. Pair-wise comparisons of tilapia TNF with other teleost sequences indicated its close relation with TNF identified from other spiny-rayed fish species. To determine the evolutionary position of tilapia TNF, phylognetic analysis were done using various methods. Independent analysis using different methods yielded similar trees indicating the clustering of tilapia TNF with TNF from other spiny-rayed fish. Meanwhile a parallel evolution of TNF can be observed between teleost and mammalian TNF (Fig. 3). 3.2. Transcriptional regulation of tilapia TNF Constitutive expression of tilapia TNF was observed in purified NCC from tilapia tissues (Fig. 4a). Expression of TNF in various tissues was verified by RT-PCR suggesting the ubiquitous nature of its expression as reported for other species (Fig. 4b). The TNF gene transcription was not upregulated in NCC upon treatment with LPS or PMA/Ca2+ ionophore (data not shown). However, exposing tilapia to acute stress treatment was found to upregulate the transcription of TNF in NCC (Fig. 4c). We have previously shown the enhancement of NCC activity upon exposure to stress-activated serum factors (Bishop et al., 2000, Jaso-Friedmann et al., 2000b, Ruiz et al., 2001). 201 Similarly, cDNA from catfish NCC was used to verify the expression of TNF-alpha in those cells using primer pairs designed based on the TNF-alpha sequence published (NCBI accession number: AJ417565) for that species (data not shown). 3.3. Production of recombinant tilapia TNF Recombinant tilapia TNF was expressed in E. coli using pET-21b expression vector. The mature form of tilapia TNF with a His-Tag at the N-terminal was expressed at high levels upon induction with IPTG. The recombinant protein was purified using Nickel affinity chromatography (Fig. 5a and b). The reactivity of commercially available anti-TNF antibodies towards recombinant tilapia TNF was assessed by Western blot analysis. Among those antibodies, a polyclonal antibody developed against murine TNF was cross-reactive with this protein (data not shown). 3.4. Tilapia NCC constitutively express membrane-bound form of TNF Expression of membrane-bound form of TNF by NCC was determined by flowcytometry. NCC were purified from peripheral blood and non-permeabilized cells were stained with the polyclonal anti-TNF antibody and secondary antibody labeled with FITC. Expression of TNF on the surface was verified by one and two-color analysis. Control rabbit IgGs and conjugate controls were used to discriminate between non-specific binding of antibodies to the cells (Fig. 6a). Approximately 50% of cells were double positive for TNF antibody binding and 5C6 (monoclonal antibody binding to NCCRP-1, an activation marker on NCC). Similar analysis using NCC purified from other tissues yielded similar results. 202 Figure 6a shows a population of NCCRP-1 positive cells that is negative for surface expression of TNF (single positive population). We have detected the presence of lower molecular weight forms of TNF (secreted/mature form) by Western blot analysis of tilapia NCC lysates (data not shown). The possibility that repeated handling of the cells during the purification steps could have resulted in the activation of NCC and release of the soluble form of TNF-alpha resulting in the single positive cell population was investigated. Leukocytes were purified in the presence of sodium azide (to prevent secretion) and analyzed for the surface expression of TNF-alpha. Under these conditions, anti-TNF polyclonal antibody bound to 90% of purified NCC. The specificity of staining with the anti-TNF antibody was verified by adsorption of the antibody with Ni-NTA- immobilized recombinant tilapia TNF (Fig. 6b). Staining of NCC with the adsorbed antibody was reduced. 3.5. Tilapia TNF can induce cytotoxicity in susceptible targets WEHI-164S cells are commonly used to assess the TNF bioactivity. Recombinant tilapia TNF induced dose-dependant cytotoxicity of these cells in the presence of actinomycin D. As expected, the cytotoxicity induced by murine TNF was significantly higher than that of the tilapia TNF, and was even apparent in the absence of actinomycin D (Fig. 7a). Actinomycin D treatment completely abolished the cytotoxicity caused by fish TNF. Similar results were reported for recombinant TNF from other fish species (Garcia-Castillo et al., 2004). Tilapia NCC were also effective in killing actinomycin D-treated WEHI target cells. The specificity of the killing was demonstrated in inhibition experiments using anti-TNF polyclonal antibody to block NCC cytotoxicity (Fig. 7b). 203 3.6. Recombinant tilapia TNF protects NCC from activation-induced cell death We have previously reported the regulation of activation-induced cell death in NCC by stress-activated serum factors (Bishop et al., 2000, Evans et al., 2001). The role of the secreted form of TNF in this process was assessed by incubating NCC in the presence of increasing concentrations of recombinant TNF followed by measurement of DNA hypoploidy. Recombinant TNF appears to have a dose-dependent protective effect on NCC. Maximum protection was achieved at 100 ng/ml of the recombinant protein, and diminished at higher concentrations (Fig. 8). To demonstrate the specificity of this action, recombinant TNF was treated with anti-TNF polyclonal antibody and immunoprecipitated before adding the supernatants to the purified NCC. Depletion of recombinant TNF with the antibody diminished the protective effect (Fig. 8). 3.7. Recombinant TNF upregulates transcription of granzyme in tilapia NCC The partial sequence of a granzyme expressed in tilapia NCC has already been reported (Praveen et al., 2004). Primers were designed based on that sequence and semi-quantitative RTPCR was used to measure the granzyme gene transcription upon treatment of tilapia NCC with recombinant TNF. A very significant increase in the transcription of granzyme gene was observed within two hours of treatment, indicating a possible role of TNF in enhancing the cellmediated cytotoxicity induced by NCC (Fig. 9). 4. DISCUSSION In this study, we present the molecular cloning of TNF from nonspecific cytotoxic cells of teleost fish and we suggest its role in the effector functions of those cells. The transcript has a 204 long 3’ untranslated region with multiple RNA instability motifs (Fig. 1), which are particularly prevalent among mRNAs encoding many cytokines and other proteins involved in inflammation (Caput et al., 1986). The deduced tilapia TNF protein showed highest degree of amino acid homology with TNF alpha from other spiny-rayed fish. Various sequence analysis algorithms suggested a type II transmembrane topology for tilapia TNF, giving rise to the possibility of membrane bound as well as secreted forms. In mammals the conversion of membrane-bound to the soluble form of TNF is achieved by a metalloproteinase called TNF-alpha converting enzyme (Blobel, 1997). Multiple sequence alignment analysis of the tilapia TNF revealed the conservation of the Thr-Leu motif which is associated with the protease processing of TNF. This would suggest that TNF from NCC is processed by a similar enzyme as in the case of mammalian TNF. Although tilapia TNF has a signature domain structure that is conserved among other members of the family, the deviations from mammalian TNF sequences are well in agreement with other fish TNF sequences (Fig. 2a). The cysteine residues responsible for the conserved tertiary structure among TNF alpha sequences as well as many other C-terminal residues are conserved in tilapia TNF. The molecular modeling studies predicted a -jellyroll structure for tilapia TNF (Fig. 2b) similar to human TNF-alpha (Gruss, 1996, Idriss and Naismith, 2000b). The results obtained thus far indicate that fish have only TNF-alpha. This argument is strengthened by the discovery of this molecule in tilapia which also has more similarity to TNFalpha sequences. Conservation of two cysteine residues crucial in correct folding as well as the lack of similarity of residues surrounding the point of cleavage, suggest that the tilapia TNF is more closely related to mammalian TNF-alpha than to TNF-beta. Phylogenetic analysis of 205 members of the TNF superfamily indicated the clustering of tilapia TNF with other TNF-alpha sequences (Fig. 3). In mammals, TNF is produced by macrophages, monocytes, neutrophils, NK cells and T cells. We report expression studies using RT-PCR analysis that indicate the constitutive expression of tilapia TNF in all tissues examined (Fig. 4b), including purified NCC. High level of expression of TNF in NCC was verified by RT-PCR (Fig. 4a). Evidence for the expression of TNF in channel catfish cytotoxic cell-line has been presented before (Zou et al., 2003b). Reports on transcriptional upregulation of TNF are highly variable among various fish species. TNFalpha expression was up-regulated by stimulating isolated primary leukocytes of flounder and trout with LPS, PMA, recombinant IL-1 or ConA (Hirono et al., 2000, Laing et al., 2001). However, TNF is constitutively expressed in all tissues examined in seabream. Both LPS and MAF failed to increase the TNF gene expression in cultured seabream macrophages (GarciaCastillo et al., 2002). Treatment of tilapia NCC with LPS or PMA/Ca2+ ionophore also failed to upregulate TNF expression (data not shown). However, there was an increase in the expression of TNF with treatment of NCC with stress-activated serum from tilapia (Fig. 4c). We have previously demonstrated the role of stress-activated serum factors (SASF) in the regulation of NCC functions (Bishop et al., 2000, Jaso-Friedmann et al., 2000b, Ruiz et al., 2001). Upregulation of TNF in NCC by SASF is indirect evidence for the role of TNF in upregulation of cytotoxicity induced by NCC purified from stressed fish. Biological activities of recombinant TNF from rainbow trout and seabream have previously been reported (Garcia-Castillo et al., 2004, Zou et al., 2003a). However, membrane expression of TNF has not been reported in any population of fish cells. Tilapia TNF was expressed as a recombinant form in bacteria using pET-21b expression vector. The protein was 206 purified using metal affinity chromatography (Fig. 5a&b) and cross-reactivity of an anti-murine TNF polyclonal antibody with the fish protein was verified by Western blot analysis. This antibody was used for staining of membrane TNF on NCC using one and two-color flowcytometry analysis (Fig. 6a). The specificity of the antibody was verified by depleting the antibody with recombinant tilapia TNF, immobilized on Ni-NTA agarose beads (Fig. 6b). These results provided evidence for the presence of the cell surface form of TNF on fish NCC, which demonstrates the conserved expression patterns as compared to mammalian cells. Expression of the membrane-bound form of TNF on mammalian NK cells, macrophages and CTL and its role in the cytotoxic functions of these cells have been previously demonstrated (Caron et al., 1999, Monastra et al., 1996). The murine fibrosarcoma cell line WEHI is generally used to assess mammalian TNF cytotoxicity (Austgulen et al., 1986, Espevik and Nissen-Meyer, 1986). The species specificity of TNF in induction of cytotoxicity has been previously reported (Lewis et al., 1991) and this is in agreement with a recent report where recombinant seabream TNF-alpha failed to induce cytotoxicity in L929 cells (Garcia-Castillo et al., 2004). In contrast, we now report that recombinant tilapia TNF induced cytotoxicity of WEHI cells in the presence of actinomycin D, though to a lesser extent than murine TNF-alpha (Fig. 7a). We propose that the discrepancies between our results with tilapia rTNF and other fish TNF may be attributed to the use of different apoptosis detection method. We used DNA hypoploidy, which appears to be a more sensitive assay for WEHI cells than those previously used. The ability of NCC to kill TNF sensitive cells was demonstrated with chromium release assays using WEHI cells as targets. The specific role of TNF in this experiment was demonstrated by neutralizing the killing with 207 addition of anti-TNF antibodies (Fig. 7b). These results suggest a critical role of TNF in cellmediated cytotoxicity in teleosts. The role of human recombinant TNF-alpha in negative regulation of T cell response has been documented (Sarin et al., 1995a). In human T cells TNF-alpha as well as LT were upregulated only upon activation (Gehr et al., 1992). We have previously reported that NCC undergo activation induced cell death, which is regulated by SASF (Bishop et al., 2000, Evans et al., 2001). However, the actual mediators of this regulation were not identified. Incubation of purified NCC with recombinant TNF resulted in increased survival from cell death, indicating a protective mechanism (Fig. 8). The better protection at lower concentrations of recombinant TNF could be explained by a concentration effect on affinity to bind to the two TNF receptors. However, the expression of TNF receptors on tilapia NCC has not been demonstrated so far. Using flowcytometry of non-permeabilized NCC, we have demonstrated that these cells constitutively express membrane bound TNF-alpha (Fig. 6). Similarly, lower molecular weight forms (mature, released form) of TNF were identified by Western blot analysis of activated NCC lysates (data not shown). These results suggested the presence of both, membrane bound and secreted TNF in tilapia NCC. Although there appears to be a marked difference in the mode of action between the two forms of mammalian TNF ligand superfamily members, identification of a more susceptible cell line for tilapia TNF would be necessary to dissect out the difference in activities of the two forms of TNF expressed on NCC. TNF-alpha is the most important pro-inflammatory cytokine released by many cells during immune responses. The autocrine/paracrine growth factor effect of TNF on T cells has been reported. Activation of T cells with TNF induces the expression of TNF receptors and secretion of cytokines (Scheurich et al., 1987, Sung et al., 1988). Lymphokine-activated killer 208 cells and CTL are optimally induced with TNF, in combination with IL-2, resulting in an increase in CTL granule constituents like granzyme A (Owen-Schaub et al., 1989, Robinet et al., 1990). These studies suggested the importance of TNF in the regulation of cytotoxic effector functions. We have previously reported the expression of a granzyme-like protease in tilapia NCC (Praveen et al., 2004). Now we show that treatment of tilapia NCC with recombinant TNF resulted in transcriptional upregulation of granzyme-like protease in those cells (Fig. 9). Thus, a mechanism of regulation of cytotoxicity by TNF similar to that in mammals is suggested in NCC. Our results demonstrated the constitutive expression of both membrane-bound as well as secreted forms of TNF-alpha in the cytotoxic cells of a teleost. The recombinant analog could induce target cell death of a mammalian cell line, to a lesser extent than its murine counterpart. Recombinant TNF was also effective in protecting NCC from cell death and up-regulating effector molecules of cytotoxicity. By analogy to the mammalian system, TNF-alpha can be expected to play a significant role in the regulation of cytotoxic cell functions in teleosts. ACKNOWLEDGEMENTS We acknowledge the assistance of Mr. John Leary, Dr. Neelesh Sharma, Dr. Jayakumar Poovassery, Dr. Quanren He and Dr. Brian McNeal at the College of Veterinary Medicine, University of Georgia for providing valuable suggestions, reagents and facilities. Research was supported by BARD # 10-21-RR211-236 and Veterinary Medical Experimental Station Grants at the College of Veterinary Medicine, University of Georgia. 209 REFERENCES Adams, J.M., Cory, S., 1998. The Bcl-2 protein family: arbiters of cell survival. Science 281, 1322-1326. Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, Z., Miller, W., Lipman, D.J., 1997. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389-3402. Ashkenazi, A., 2002. Targeting death and decoy receptors of the tumour-necrosis factor superfamily. Nat. Rev. Cancer 2, 420-430. Austgulen, R., Kildahl-Andersen, O., Espevik, T., 1986. Monocyte-mediated drug-dependent cellular cytotoxicity: effects on different WEHI 164 target cell lines. Cancer Immunol. Immunother. 22, 176-180. Bishop, G.R., Jaso-Friedmann, L., Evans, D.L., 2000. Activation-induced programmed cell death of nonspecific cytotoxic cells and inhibition by apoptosis regulatory factors. Cell Immunol. 199, 126-137. Bishop, G.R., Taylor, S., Jaso-Friedmann, L., Evans, D.L., 2002. Mechanisms of nonspecific cytotoxic cell regulation of apoptosis: cytokine-like activity of Fas ligand. Fish. Shellfish. Immunol. 13, 47-67. Blobel, C.P., 1997. Metalloprotease-disintegrins: links to cell adhesion and cleavage of TNF alpha and Notch. Cell 90, 589-592. Caput, D., Beutler, B., Hartog, K., Thayer, R., Brown-Shimer, S., Cerami, A., 1986. Identification of a common nucleotide sequence in the 3'-untranslated region of mRNA molecules specifying inflammatory mediators. Proc. Natl. Acad. Sci. U. S. A 83, 16701674. Carlson, R.L., Evans, D.L., Graves, S.S., 1985. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). V. Metabolic requirements of lysis. Dev. Comp Immunol. 9, 271-280. Caron, G., Delneste, Y., Aubry, J.P., Magistrelli, G., Herbault, N., Blaecke, A., Meager, A., Bonnefoy, J.Y., Jeannin, P., 1999. Human NK cells constitutively express membrane TNF-alpha (mTNFalpha) and present mTNFalpha-dependent cytotoxic activity. Eur. J. Immunol. 29, 3588-3595. Eissner, G., Kohlhuber, F., Grell, M., Ueffing, M., Scheurich, P., Hieke, A., Multhoff, G., Bornkamm, G.W., Holler, E., 1995. Critical involvement of transmembrane tumor necrosis factor-alpha in endothelial programmed cell death mediated by ionizing radiation and bacterial endotoxin. Blood 86, 4184-4193. Espevik, T., Nissen-Meyer, J., 1986. A highly sensitive cell line, WEHI 164 clone 13, for measuring cytotoxic factor/tumor necrosis factor from human monocytes. J. Immunol. Methods 95, 99-105. 210 Evans, D.L., Carlson, R.L., Graves, S.S., Hogan, K.T., 1984. Nonspecific cytotoxic cells in fish (Ictalurus punctatus). IV. Target cell binding and recycling capacity. Dev. Comp Immunol. 8, 823-833. Evans, D.L., Leary, J.H., III, Jaso-Friedmann, L., 1998. Nonspecific cytotoxic cell receptor protein-1: a novel (predicted) type III membrane receptor on the teleost equivalent of natural killer cells recognizes conventional antigen. Cell Immunol. 187, 19-26. Evans, D.L., Leary, J.H., III, Jaso-Friedmann, L., 2001. Nonspecific cytotoxic cells and innate immunity: regulation by programmed cell death. Dev. Comp Immunol. 25, 791-805. Faisal, M., Ahmed, I.I., Peters, G., Cooper, E.L., 1989. Natural Cyto-Toxicity of Tilapia Leukocytes. Diseases of Aquatic Organisms 7, 17-22. Fesik, S.W., 2000. Insights into programmed cell death through structural biology. Cell 103, 273-282. Garcia-Castillo, J., Chaves-Pozo, E., Olivares, P., Pelegin, P., Meseguer, J., Mulero, V., 2004. The tumor necrosis factor alpha of the bony fish seabream exhibits the in vivo proinflammatory and proliferative activities of its mammalian counterparts, yet it functions in a species-specific manner. Cell Mol. Life Sci. 61, 1331-1340. Garcia-Castillo, J., Pelegrin, P., Mulero, V., Meseguer, J., 2002. Molecular cloning and expression analysis of tumor necrosis factor alpha from a marine fish reveal its constitutive expression and ubiquitous nature. Immunogenetics 54, 200-207. Gehr, G., Gentz, R., Brockhaus, M., Loetscher, H., Lesslauer, W., 1992. Both tumor necrosis factor receptor types mediate proliferative signals in human mononuclear cell activation. J. Immunol. 149, 911-917. Goetz, F.W., Planas, J.V., Mackenzie, S., 2004. Tumor necrosis factors. Dev. Comp Immunol. 28, 487-497. Graves, S.S., Evans, D.L., Dawe, D.L., 1985. Antiprotozoan activity of nonspecific cytotoxic cells (NCC) from the channel catfish (Ictalurus punctatus). J. Immunol. 134, 78-85. Green, D.R., 2000. Apoptotic pathways: paper wraps stone blunts scissors. Cell 102, 1-4. Greenlee, A.R., Brown, R.A., Ristow, S.S., 1991. Nonspecific cytotoxic cells of rainbow trout (Oncorhynchus mykiss) kill YAC-1 targets by both necrotic and apoptic mechanisms. Dev. Comp Immunol. 15, 153-164. Grell, M., Douni, E., Wajant, H., Lohden, M., Clauss, M., Maxeiner, B., Georgopoulos, S., Lesslauer, W., Kollias, G., Pfizenmaier, K., ., 1995a. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83, 793-802. 211 Grell, M., Douni, E., Wajant, H., Lohden, M., Clauss, M., Maxeiner, B., Georgopoulos, S., Lesslauer, W., Kollias, G., Pfizenmaier, K., ., 1995b. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83, 793-802. Gruss, H.J., 1996. Molecular, structural, and biological characteristics of the tumor necrosis factor ligand superfamily. Int. J. Clin. Lab Res. 26, 143-159. Hirono, I., Nam, B.H., Kurobe, T., Aoki, T., 2000. Molecular cloning, characterization, and expression of TNF cDNA and gene from Japanese flounder Paralychthys olivaceus. J. Immunol. 165, 4423-4427. Hunt, A., Evan, G., 2001. Apoptosis. Till death us do part. Science 293, 1784-1785. Idriss, H.T., Naismith, J.H., 2000a. TNF alpha and the TNF receptor superfamily: structurefunction relationship(s). Microsc. Res. Tech. 50, 184-195. Idriss, H.T., Naismith, J.H., 2000b. TNF alpha and the TNF receptor superfamily: structurefunction relationship(s). Microsc. Res. Tech. 50, 184-195. Jaso-Friedmann, L., Evans, D.L., 1999. Mechanisms of cellular cytotoxic innate resistance in tilapia (Oreochromis nilotica). Dev. Comp Immunol. 23, 27-35. Jaso-Friedmann, L., Leary, J.H., III, Evans, D.L., 2000a. Role of nonspecific cytotoxic cells in the induction of programmed cell death of pathogenic protozoans: participation of the Fas ligand-Fas receptor system. Exp. Parasitol. 96, 75-88. Jaso-Friedmann, L., Peterson, D.S., Gonzalez, D.S., Evans, D.L., 2002. The antigen receptor (NCCRP-1) on catfish and zebrafish nonspecific cytotoxic cells belongs to a new gene family characterized by an F-box-associated domain. J. Mol. Evol. 54, 386-395. Jaso-Friedmann, L., Ruiz, J., Bishop, G.R., Evans, D.L., 2000b. Regulation of innate immunity in tilapia: activation of nonspecific cytotoxic cells by cytokine-like factors. Dev. Comp Immunol. 24, 25-36. Kashii, Y., Giorda, R., Herberman, R.B., Whiteside, T.L., Vujanovic, N.L., 1999. Constitutive expression and role of the TNF family ligands in apoptotic killing of tumor cells by human NK cells. J. Immunol. 163, 5358-5366. Kwon, B., Youn, B.S., Kwon, B.S., 1999. Functions of newly identified members of the tumor necrosis factor receptor/ligand superfamilies in lymphocytes. Curr. Opin. Immunol. 11, 340-345. Laing, K.J., Wang, T., Zou, J., Holland, J., Hong, S., Bols, N., Hirono, I., Aoki, T., Secombes, C.J., 2001. Cloning and expression analysis of rainbow trout Oncorhynchus mykiss tumour necrosis factor-alpha. Eur. J. Biochem. 268, 1315-1322. 212 Lewis, M., Tartaglia, L.A., Lee, A., Bennett, G.L., Rice, G.C., Wong, G.H., Chen, E.Y., Goeddel, D.V., 1991. Cloning and expression of cDNAs for two distinct murine tumor necrosis factor receptors demonstrate one receptor is species specific. Proc. Natl. Acad. Sci. U. S. A 88, 2830-2834. Locksley, R.M., Killeen, N., Lenardo, M.J., 2001. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104, 487-501. McGeoch, D.J., 1985. On the predictive recognition of signal peptide sequences. Virus Res. 3, 271-286. McKinney, E.C., Schmale, M.C., 1994. Damselfish with neurofibromatosis exhibit cytotoxicity toward tumor targets. Dev. Comp Immunol. 18, 305-313. Monastra, G., Cabrelle, A., Zambon, A., Rosato, A., Macino, B., Collavo, D., Zanovello, P., 1996. Membrane form of TNF alpha induces both cell lysis and apoptosis in susceptible target cells. Cell Immunol. 171, 102-110. Owen-Schaub, L.B., Crump, W.L., III, Morin, G.I., Grimm, E.A., 1989. Regulation of lymphocyte tumor necrosis factor receptors by IL-2. J. Immunol. 143, 2236-2241. Praveen, K., Evans, D.L., Jaso-Friedmann, L., 2004. Evidence for the existence of granzyme-like serine proteases in teleost cytotoxic cells. J. Mol. Evol. 58, 449-459. Robinet, E., Branellec, D., Termijtelen, A.M., Blay, J.Y., Gay, F., Chouaib, S., 1990. Evidence for tumor necrosis factor-alpha involvement in the optimal induction of class I allospecific cytotoxic T cells. J. Immunol. 144, 4555-4561. Ruiz, J., Leary, J.H., III, Jaso-Friedmann, L., 2001. Phosphorylation-induced activation of tilapia nonspecific cytotoxic cells by serum cytokines. Dis. Aquat. Organ 46, 129-137. Saeij, J.P., Stet, R.J., de Vries, B.J., van Muiswinkel, W.B., Wiegertjes, G.F., 2003. Molecular and functional characterization of carp TNF: a link between TNF polymorphism and trypanotolerance? Dev. Comp Immunol. 27, 29-41. Sarin, A., Conan-Cibotti, M., Henkart, P.A., 1995a. Cytotoxic effect of TNF and lymphotoxin on T lymphoblasts. J. Immunol. 155, 3716-3718. Sarin, A., Conan-Cibotti, M., Henkart, P.A., 1995b. Cytotoxic effect of TNF and lymphotoxin on T lymphoblasts. J. Immunol. 155, 3716-3718. Scheurich, P., Thoma, B., Ucer, U., Pfizenmaier, K., 1987. Immunoregulatory activity of recombinant human tumor necrosis factor (TNF)-alpha: induction of TNF receptors on human T cells and TNF-alpha-mediated enhancement of T cell responses. J. Immunol. 138, 1786-1790. Schneider, P., Holler, N., Bodmer, J.L., Hahne, M., Frei, K., Fontana, A., Tschopp, J., 1998. Conversion of membrane-bound Fas(CD95) ligand to its soluble form is associated with 213 downregulation of its proapoptotic activity and loss of liver toxicity. J. Exp. Med. 187, 1205-1213. Singer, S.J., 1990. The structure and insertion of integral proteins in membranes. Annu. Rev. Cell Biol. 6, 247-296. Suda, T., Hashimoto, H., Tanaka, M., Ochi, T., Nagata, S., 1997. Membrane Fas ligand kills human peripheral blood T lymphocytes, and soluble Fas ligand blocks the killing. J. Exp. Med. 186, 2045-2050. Sung, S.S., Bjorndahl, J.M., Wang, C.Y., Kao, H.T., Fu, S.M., 1988. Production of tumor necrosis factor/cachectin by human T cell lines and peripheral blood T lymphocytes stimulated by phorbol myristate acetate and anti-CD3 antibody. J. Exp. Med. 167, 937953. Tanaka, M., Itai, T., Adachi, M., Nagata, S., 1998. Downregulation of Fas ligand by shedding. Nat. Med. 4, 31-36. von Heijne G., 1986. A new method for predicting signal sequence cleavage sites. Nucleic Acids Res. 14, 4683-4690. Wallach, D., Varfolomeev, E.E., Malinin, N.L., Goltsev, Y.V., Kovalenko, A.V., Boldin, M.P., 1999. Tumor necrosis factor receptor and Fas signaling mechanisms. Annu. Rev. Immunol. 17, 331-367. Zou, J., Peddie, S., Scapigliati, G., Zhang, Y., Bols, N.C., Ellis, A.E., Secombes, C.J., 2003a. Functional characterisation of the recombinant tumor necrosis factors in rainbow trout, Oncorhynchus mykiss. Dev. Comp Immunol. 27, 813-822. Zou, J., Secombes, C.J., Long, S., Miller, N., Clem, L.W., Chinchar, V.G., 2003b. Molecular identification and expression analysis of tumor necrosis factor in channel catfish (Ictalurus punctatus). Dev. Comp Immunol. 27, 845-858. 214 Figure 6.1. Nucleotide and deduced amino acid sequence of tilapia TNF. Start and stop codons are represented in bold letters. The predicted transmembrane domain is highlighted and ATTTA motifs are bold with underline. Polyadenylation signal and poly-A tail are italicized. 215 1 51 101 151 201 251 301 351 401 451 501 551 601 651 701 751 801 851 901 951 1001 1051 1101 1151 1201 1251 1301 ACTCTTAACACAAACAGCACACTCAAAGAGAGAAGTGTCAGCACAGAAGA CACAGACAACTGTTCGGTTTCTGGATTTCTTTAAAGAGCAGACAGCAGAG M V A Y T T T P V D V E GCTTTGTGCACAGCATGGTTGCATACACAACCACACCAGTTGACGTCGAG A G P E A K T V V L V E K K S P A GCTGGTCCTGAGGCGAAGACTGTAGTTTTAGTTGAGAAGAAATCACCTGC E W I W K V C A V L V V V A L C AGAGTGGATATGGAAAGTGTGCGCCGTCCTCGTCGTCGTGGCTCTTTGTT L A G V L L F A W Y W N T R P E R TAGCAGGTGTCCTGCTGTTTGCCTGGTACTGGAATACAAGGCCAGAAAGG M T Q L G Q P E A L K A K N T G D ATGACACAATTAGGACAGCCAGAAGCACTAAAGGCGAAGAACACTGGCGA K T E P H S T L K R I S S K A K CAAAACAGAGCCCCACTCCACGCTAAAACGGATCAGCAGCAAAGCCAAGG A A I H L E G S D S K G H L E W R CAGCCATCCATCTAGAAGGCAGCGACTCAAAAGGTCATCTGGAGTGGAGG N G Q G Q A F A Q G G F K L E A N AATGGTCAAGGCCAGGCGTTTGCTCAGGGCGGCTTCAAGCTGGAGGCCAA K I I I P H T G L Y F V Y S Q A TAAAATCATCATCCCACACACTGGCCTGTACTTCGTCTACAGCCAGGCGT S F R V I C G N T D E N E D E E K CCTTCAGGGTGATCTGCGGGAATACTGACGAAAATGAAGATGAGGAAAAA S L T I L S H R I W R Y S E S M G AGCCTCACAATTCTCAGCCACAGGATCTGGCGCTACTCAGAGTCTATGGG S S S T L M S A L R S A C Q D T AAGCAGCTCCACTCTGATGAGCGCCCTGAGGTCGGCGTGCCAAGACACCA I Q D S F S D H G W Y N A I Y L G TTCAGGATAGCTTCTCAGACCACGGCTGGTACAACGCCATTTACCTGGGC A V F Q L N E G D T L W T E T N Q GCTGTGTTTCAGCTGAACGAAGGAGACACGCTGTGGACGGAAACCAACCA L S E L E T D E G R T F F G V F GCTATCAGAGCTGGAGACTGACGAAGGAAGGACCTTCTTTGGCGTGTTTG A L * CACTTTGAAATGACTCTCTGCTTTGAGCAGAAAAGCTGCACAGAGCCAGA TGTTTTGGCTTAAAAACTATGTGTATATATAGTTTTTTATTTCATCCACA TGGTGATGTTTAAATATTGAAATCTCAATGGAGATGAAATTTCTAGCTGA AAACGCTAGGCTGATTTTTGAAAACTATATAAACTGTAACAGTTTGCAAC ATTGTTTCTACTTTAGGCACTTTTGACATATTATTTATACTGACCTGAGA TTGTATTAGGCTAGATTTCCTCTGCTGTATCAGATGATGACGTAGCTCTT TGGCTCAAGAGTTAAGATACAGAGGTGCTGTCATAGCGTGTATGTATTTA TTTGTATTTATTTGTATTTAAATCACTGGGACCGGGGCTTTAAAAGTATT TATATAAGTTTTGAAAGGAATAAAAATGCAGAAAACTACAAAAAAAAAAA AAAAAA 216 12 29 45 62 79 95 112 129 145 162 179 195 212 229 245 247 Figure 6.2. (a) Multiple sequence alignment of tilapia TNF with other teleosts and mammalian TNF sequences. The analysis was performed using Clustal W provided with vector NTI package, version 6 (InforMax Inc). Darker shading represents identical or residues with similar properties in 100% of the sequences and lighter shading represents identical or residues with similar properties in 80% or more of all the sequences. (b) Predicted 3D model of mature tilapia TNF monomer in comparison to human TNF. Models were generated using SWISS-MODEL in the first approach mode accessible via the internet (http://www.expasy.org/swissmod). 217 A Tilapia TNF AY428948 Pagrus TNF-a AAP76392 Sparus TNF-a Q8JFG3 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF8633 Trout TNF-a-1 CAB89521 Danio AAR06286 Catfish TNF-a CAD10389 Carp TNF-a-1 CAC84641 Human TNF-a AAA61198 Mouse TNF-a NP-038721 Rat TNF-a NP-036807 : : : : : : : : : : : : -MVAYTTTPVDVEAG-----------PEAKTVVLVEKKSPAEWIWKVCAVLVVVALCLAG -------------------------------------------MWKLSVTLLVAALCFGG -MGAYTTAPCDLEMG-----------PEERTVVLIEKKSSTGWMWKVSVALLIAALCFAG -------------------------------------------MCKVLGGLFIVALCLGG -MEGYAMTTGDMERGLENSLVDSGPVYKTTVTAVAERKASRGWLWRLCGVLLVAALCAAA -MEGYAMTPEDMER---------GPVYNTTVTAVAEGKASRGWLWRLCGVLLIAGLCAAA -MKLESRAFLDVEEGE--------LPLPLVMVSRRKAGSSKSGVWRVFGTILAVGLCAAA -MASDSQVVLDVD-------------GPRVTIVREKASWSSSGVWRTCGVLLAVALCAAA MMDLESQLLEEG--GL--------LPLPQVMVSRRKSGSSKSGVWRVCGVLLAVALCAAA --MSTESMIRDVELA--------------EEALPKKTGGPQGSRRCLFLSLFSFLIVAGA --MSTESMIRDVELA--------------EEALPQKMGGFQNSRRCLCLSLFSFLLVAGA --MSTESMIRDVELA--------------EEALPKKMGGLQNSRRCLCLSLFSFLLVAGA : : : : : : : : : : : : 48 17 48 17 59 50 51 46 50 44 44 44 Tilapia TNF AY428948 Pagrus TNF-a AAP76392 Sparus TNF-a Q8JFG3 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF8633 Trout TNF-a-1 CAB89521 Danio AAR06286 Catfish TNF-a CAD10389 Carp TNF-a-1 CAC84641 Human TNF-a AAA61198 Mouse TNF-a NP-038721 Rat TNF-a NP-036807 : : : : : : : : : : : : VLLFAWYWNTRPERMTQLGQPEALKAKNTGDKTEPHSTLKRISSKAKAAIHLEG--SDSK VLLFAWYWNGKPQTLIQSGQTEALTKTDTAEKTDPHSTLRRISSKAKAAIHLEGSYDEDE VLLFAWYWNGKPEILIHSGQSEALTKKDHAEKTDPHSTLKRISSKAKAAIHLEGSYDEDE VLAFSWYTN-KSEMMTQSGQTAALSQKDCAEKTEPHNTLRQISSRAKAAIHLEGRDEEDE ALLFAWCQHGRLETMQDGMEP---QLEILIGAKDTHHTLKQIAGNAKAAIHLEG--EYNP ALLFAWCQHGRPSTMQDEIEP---QLEILIGAKDTHHTLKQIAGNAKAAIHLEG--EYNP AVCFTLHKT-------QGNQQ----DGS-VLRLTLRDRISQGNFTSKAAIHLTGGYN--S AVCFSQNKT-------HNKPD----ETQ-EIKHSLRQ-IS---QTAKAAIHLSGHYNPQV AVCFTLNKS-------QNNQE----GGN-ALRLTLRDHLSKANVTSKAAIHLIGAYEPKV TTLFCLLHFGVIGPQREE-SPRDLSLISPLAQAVRS---SSRTPSDKPVAHVVAN----TTLFCLLNFGVIGPQRDEKFPNGLPLISSMAQTLTLR-SSSQNSSDKPVAHVVAN----TTLFCLLNFGVIGPNKEEKFPNGLPLISSMAQTLTLR-SSSQNSSDKPVAHVVAN----- : : : : : : : : : : : : 106 77 108 76 114 105 97 90 98 95 98 98 Tilapia TNF AY428948 Pagrus TNF-a AAP76392 Sparus TNF-a Q8JFG3 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF8633 Trout TNF-a-1 CAB89521 Danio AAR06286 Catfish TNF-a CAD10389 Carp TNF-a-1 CAC84641 Human TNF-a AAA61198 Mouse TNF-a NP-038721 Rat TNF-a NP-036807 : : : : : : : : : : : : G----HLEWRNGQGQAFAQGGFKLEANKIIIPHTGLYFVYSQASFRVI-CGNTDEN--ED GSKD-QVGWKSGQGQAFAQGGFRLVDNKIVIPQTGLYFVYSQASFRVS-CSDGEE---EG GLKD-QVEWKNGQGQAFAQGGFRLVDNKIVIPHTGLYFVYSQASFRVS-CSDGDE---EG ETSENKLVWKNDEGLAFTQGGFELVDNHIIIPRSGLYFVYSQASFRVSCSSDDADDGKEA NLTADTVQWRKDDGQAFSQGGFKLQGNQILIPHTGLFFVYSQASFRVK-CNGPGE----NLSADTVQWRKDDGQAFSQGGFELQGNQILIPHTGLFFVYSQASFRVK-CNSPGE----ESK--TLDWRDDQDQAFSSGGLKLVNREIIIPDDGIYFVYSQVSLHIS-CTSELTEEQ-SSV--SMQWFDNADQSFSSG-LKLEDNEIKILRDGLYFVYSQASYRLL-CKAEGDETEGE STE--TLDWKKNQDQAFTSGGLKLVEREIIIPTDGIYFVYSQVSFHIN-CKTNMTEDHDPQAEGQLQWLNRRANALLANGVELRDNQLVVPSEGLYLIYSQVLFKGQGCPS-------HQVEEQLEWLSQRANALLANGMDLKDNQLVVPADGLYLVYSQVLFKGQGCP--------HQAEEQLEWLSQRANALLANGMDLKDNQLVVPADGLYLIYSQVLFKGQGCP--------- : : : : : : : : : : : : 159 132 163 136 168 159 152 146 154 147 149 149 Tilapia TNF AY428948 Pagrus TNF-a AAP76392 Sparus TNF-a Q8JFG3 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF8633 Trout TNF-a-1 CAB89521 Danio AAR06286 Catfish TNF-a CAD10389 Carp TNF-a-1 CAC84641 Human TNF-a AAA61198 Mouse TNF-a NP-038721 Rat TNF-a NP-036807 : : : : : : : : : : : : EEKSLTILSHRIWRYSESMGSSSTLMSALRSACQDTIQDSFSDHG--WYNAIYLGAVFQL AGGHHTPLSHRISRSSESMGSDVSLMSAVRSACQNTAQDDSYSDGRGWYNTIYLGAVFQL AGRHLTPLSHRISRYSESMGSDVSLMSAVRSACQNTAQEDSYSDGRGWYNTIYLGAVFQL AEKHLTSISHRVWLFTESLGTQVSLMSAVRSACQ-KSQEDAYRDGQGWYNAIYLGAVFQL ---RTTPLSHVICRYSDSIGVNANLLSGVRSVCQQNYGNAESNIGEGWYNAVYLSAVFQL ---HTTPLSHIIWRYSDSIGVNANLLSGVRSVCQQNYGDAESEIGEGWYNAVYLGAVFQL -----VLMSHAVMRFSESYGGKKPLFSAIRSICTQEPE--S---ENLWYNTIYLGAAFHL ----VMHMSHKVSRWSDSYSSWKPLLSATRSACKKTTE--EY--QKYWYGAVYLGAAFNL ----LVHMSHTVLRYSDSYGRYMPLFSAIRTACAQASN--T---DDLWYNTIYLGAAFKL ---THVLLTHTISRIAVSYQTKVNLLSAIKSPCQRETP--EGAEAKPWYEPIYLGGVFQL ---DYVLLTHTVSRFAISYQEKVNLLSAVKSPCPKDTP--EGAELKPWYEPIYLGGVFQL ---DYVLLTHTVSRFAISYQEKVSLLSAIKSPCPKDTP--EGAELKPWYEPMYLGGVFQL : : : : : : : : : : : : 217 192 223 195 225 216 202 198 205 202 204 204 Tilapia TNF AY428948 Pagrus TNF-a AAP76392 Sparus TNF-a Q8JFG3 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF8633 Trout TNF-a-1 CAB89521 Danio AAR06286 Catfish TNF-a CAD10389 Carp TNF-a-1 CAC84641 Human TNF-a AAA61198 Mouse TNF-a NP-038721 Rat TNF-a NP-036807 : : : : : : : : : : : : NEGDTLWTETN--QLSELETDEGRTFFGVFAL NRGDRLWTETN--QLSELETEEGKTFFGVFAL NRGDKLETETN--QLSELETDEGKTFFGVFAL NEGDKLWTETN--MLSELETESGKTFFGVFAL NEGDKLWTETN--RLTDVEPEHGKNFFGVFAL NEGDKLWTETN--RLTDVEPEQGKNFFGVFAL REGDRLGTDTTTALLPMVENDNGKTFFGVFGL KAGDRLRTVMDEKLLPKVESAGGKTFFGTFSL RAGDRLRTETTEELLPSVETGDGKTFFGVFAL EKGDRLSAEINRPDYLDFAES-GQVYFGIIAL EKGDQLSAEVNLPKYLDFAES-GQVYFGVIAL EKGDLLSAEVNLPKYLDITES-GQVYFGVIAL 218 : : : : : : : : : : : : 247 222 253 225 255 246 234 230 237 233 235 235 B 219 Figure 6.3. Phylogenetic analysis of tilapia TNF: Phylogram showing relationships of tilapia TNF other TNF family members. The tree was derived by parsimony analysis, with Mega version 2. Numbers shown above the branches are bootstrap values based upon 1000 replicates for parsimony. A separate analysis using maximum likelihood and neighbor joining methods produced a tree with similar topology. The tree was rooted on a sub-tree containing mammalian fas ligand and lymphotoxin. 220 85 100 Sparus TNF-a Q8JFG3 Acanthopagrus TNF-a AAP94278 Pagrus TNF-a AAP76392 99 Tilapia TNF AY428948 97 Flounder TNF-a BAA94969 Brook Trout TNF-a AAF86331 100 100 Trout TNF-a 1 CAB89521 Catfish TNF-a CAD10389 Danio AAR06286 100 Carp TNF-a 1 CAC84641 97 Carp TNF-a 2 CAC84642 99 80 Carp TNF-a 3 BAC77690 Wallaby TNF-a O77764 99 100 Rat TNF-a NP 036807 Mouse TNF-a NP 038721 Horse TNF-a P29553 99 Human TNF-a AAA61198 84 86 100 Monkey TNF-a Q8MKG8 Mouse FasL AAB33780 Human FasL P48023 Human Lymphotoxin AAA61199 74 Mouse Lymphotoxin AAA18593 100 100 221 Rat Lymphotoxin AAA16276 Figure 6.4. Expression of tilapia TNF gene (a) RT-PCR analysis of tilapia NCC. Lib: cDNA library, cDNA: fresh cDNA from purified NCC, NTC: No Template Control. (b) RT-PCR analysis of tilapia TNF expression in various tissues. Beta-Actin was used as a control for amount and quality of RNA. AK: anterior kidney, LV: Liver, HT: heart, GL: gill, MSL: muscle, PBL: peripheral blood leukocytes. (c) Transcriptional regulation of tilapia TNF gene by stress activated serum factors (SASF). NCC were exposed to cold stress and RNA was extracted from the cells. RT-PCR analysis using beta-actin as a normalizer was used to compare the expression of TNF gene. 222 A B LV AK HT GL MSL PBL TNF β-Actin C 1.6 1.4 TNF/Actin 1.2 1 0.8 0.6 0.4 0.2 0 Control SASF 223 Figure 6.5. Purification of recombinant tilapia TNF using Ni-affinity chromatography. (a) Purification of His-tagged proteins from bacteria with pET-21-TLTNF plasmid. M: molecular weight markers, L: total lystates from the bacterial pellet, F: flow-through from the column, W1 and W2: washings of the column (buffer with 20 mM imidazole), E1-E4: first four elutions of the column (buffer with 250 nM imidazole). (b) Similar purification procedure from bacteria with empty vector. Gels were stained with Coomassie Gel Code staining reagent (Pierce, IL). 224 225 Figure 6.6. Expression of TNF on tilapia NCC membranes. (a) Two-color flowcytometric analysis of tilapia NCC using anti-TNF polyclonal antibody (followed by FITC-labeled secondary antibody) and 5C6, a monoclonal antibody detecting NCCRP-1 on NCC (followed by PE-labeled secondary antibody). (b) Single color analysis of tilapia NCC for surface expression of TNF-alpha. Tilapia NCC were purified in the presence of sodium azide and non-permeablized cells were stained for TNF. The specificity of binding was verified by treating the antibody with polyhistidine tagged recombinant tilapia TNF immobilized on Ni-NTA agarose beads or NiNTA agarose beads as a control. 226 A B 100 90 80 % Positive cells 70 60 50 40 30 20 10 0 Anti-TNF pAb Anti-TNF pAb depleted with TNF 227 Anti-TNF pAb depleted with NiNTAAgarose Figure 6.7. Cytotoxicity induced by recombinant TNF on susceptible WEHI cells. (a) WEHI cells were treated with murine or fish TNF in the presence or absence of actinomycin-D. DNA hypoploidy analysis was done to determine the cell death. (b) Cytotoxicity of WEHI cells by tilapia NCC were determined by chromium release assay. The specificity of killing was verified by blocking the killing by anti-TNF antibody. 228 A B 35 % Specific Release 30 25 Media Control 20 Control Serum Anti-TNF pAb 15 10 5 0 160 80 40 E:T Ratio 229 Figure 6.8. Recombinant TNF protects activation-induced cell death in NCC. Purified tilapia NCC were centrifuged at low speed to facilitate cell-to-cell contact and incubated in the presence or absence of recombinant tilapia TNF. Cell death after 18 hours was measured by analysis of DNA hypoploidy. Recombinant tilapia TNF (100 ng/ml) was incubated with anti-TNF polyclonal antibody or control rabbit IgGs followed by immunoprocipitation. The TNF depleted supernatants were added to the cells to verify the specificity of protection. 230 TNF IP Control Rb Igs TNF IP anti-TNF pAb TNF 6.25 ng/ml TNF 12.5 ng/ml TNF 25 ng/ml TNF 50 ng/ml TNF 100 ng/ml TNF 200 ng/ml TNF 400 ng/ml Media 0 5 10 % DNA Hypoploidy 231 15 20 Figure 6.9. Recombinant TNF upregulates the transcription of granzyme in tilapia NCC. Granzyme gene expression was analyzed by RT-PCR after treating NCC with 100 ng/ml of recombinant tilapia TNF. Results are expressed as ratios of granzyme to beta-actin, which was used as a normalizer. 232 1 0.9 0.8 TLGR/Actin 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0 0H 2H 233 4H CHAPTER 7 MOLECULAR CLONING OF CELLULAR APOPTOSIS SUSCEPTIBILITY (CAS) GENE IN OREOCHROMIS NILOTICUS AND ITS ROLE IN REGULATION OF NONSPECIFIC CYTOTOXIC CELL (NCC) FUNCTIONS1 1 Praveen, K., J. H. Leary, D. L. Evans and L. Jaso-Friedmann. 2005. Submitted to Fish and Shellfish Immunology, 02/07/2005. 234 Cellular Apoptosis Susceptibility (CAS) gene is a homologue of the Chromosome Segregation gene (CSE) in yeast [1-4] and appears to be involved in multiple cellular mechanisms. The roles that have been attributed to CAS include mitotic spindle checkpoints, cell proliferation and apoptosis [5,6]. Attenuation of CAS reduces the sensitivity of cells to TNF-α and -β mediated apoptosis, probably due to its involvement in nuclear transport of apoptosisrelated proteins [7]. CAS is highly expressed in proliferating cells but at a lower level in quiescent cells and tissues [8-12]. CAS mutants are unable to degrade cyclin B during key check points in mitosis and subsequently become arrested during cell cycle. Thus, CAS appears to play an important role in cancer development [5]. Some of the functions attributed to CAS can be partly related to its role as nuclear export factor for importin-α [13-15]. It has been demonstrated that phosphorylation of N-terminal Tyr residues in CAS by MEK-like kinases regulates the nuclear localization of CAS [8]. In normal cells, CAS is considered to function as a "switch", determining whether a cell will proliferate or undergo apoptosis [7]. Nonspecific cytotoxic cells (NCC) are the first identified cytotoxic cell population in teleosts. Since their initial description in the channel catfish [16-19], these cells have been well characterized in a number of lower vertebrates [20-25]. NCC are functionally similar to mammalian NK cells. It has been reported that human NK cells are recycled 3-4 times between different target cells in 3-h in vitro killing assays [26]. Binding of NCC to target cells (or cross linking of the activation marker nonspecific cytotoxic cell receptor protein-1, NCCRP-1 with a monoclonal antibody) has been shown to induce activation induced programmed cell death (AIPCD) in NCC, thus preventing the possibility of recycling [27,28]. NCC are active participants of the innate immune response so as 235 such, NCC must activate regulatory mechanisms of protection from AIPCD. Cells undergoing apoptosis are immunologically incompetent because they are unable to effect cytotoxic functions and they cannot secrete growth factors or cytokines necessary for regulation of immune activities [29]. One of the mechanisms of regulation of AIPCD in NCC has been identified as the soluble cytokine-like serum factors released into the serum of tilapia following acute stress (stressactivated serum factors: SASF) [30,31]. CAS was previously identified in tilapia NCC with a cross-reacting monoclonal antibody. Its expression was up-regulated in apoptosis-protected NCC treated with stress-activated serum [31]. Here we report the molecular cloning and expression of CAS in NCC, and we propose CAS expression as a mechanism of regulation of AIPCD in these cytotoxic cells. Full-length cDNA sequence of tilapia CAS (Accession number AF547173) has 3572 bp with an open reading frame (ORF) of 2916 bp. The putative polypeptide sequence encoded by this ORF consists of 971 amino acids (Fig. 1). Homology analysis of this protein using different algorithms suggested it has closest similarity to cellular apoptosis susceptibility (CAS) genes identified from various organisms. Multiple motifs with L(X)nP sequence, which is generally found in other CAS sequences are highly conserved in tilapia CAS. By searching the NCBI’s conserved domain database [32], a C-terminal CAS-CSE-1 signature motif was identified. There are two putative N-glycosylation sites (456-459 and 537-540) as well as multiple phosphorylation sites that were identified in the tilapia CAS sequence. Two of the tyrosine kinase phosphorylation sites (425-432 and 281-289) have been predicted to play an important role in regulating the movement of CAS in and out of the nucleus [8]. A basic region (positions 372-385), which is believed to be a nuclear localization or DNA binding site [2], was also found to be well conserved between fish and other CAS genes (Fig. 2). Similar to other CAS genes, 236 there is a putative Zn finger domain (His-(X)2-His-(X)6-Cys-(X)6-Cys) present in tilapia CAS at positions 613-630. Phylogenetic analysis of similar proteins identified from various species revealed these close similarities between tilapia CAS and other teleost CAS sequences (Fig. 3). Expression of CAS transcripts in various tissues was investigated by RT-PCR (Fig. 4). Higher level of expression of CAS was observed in tissues which are rich in NCC, indicating NCC as a major site of CAS expression in tilapia. A possible role of CAS in the regulation of NCC functions was identified by RT-PCR of NCC following exposure to stress-activated serum factors and recombinant tilapia TNF-alpha. We have previously reported the role of stressactivated serum components in protection of NCC from activation-induced cell death [30,31]. Transcriptional up-regulation of CAS upon exposure to stress-activated serum is an indication of the involvement of CAS as one of the mechanisms of protection against apoptotic cell death. The specific serum factors that are responsible for up-regulation of CAS transcription were not identified. In order to better understand the factors involved in AIPCD protection of NCC, experiments were next done with recombinant tilapia TNF-alpha. We have reported that this cytokine has a protective effect on NCC (Praveen et al. 2005. Molecular Immunology, In press). RT-PCR analysis of NCC exposed to recombinant tilapia TNF-alpha revealed a transcriptional up-regulation of CAS, indicating that CAS may be one of the mediators of protective effects of TNF on NCC. Although further studies are necessary to show the exact nature of the events involved in the protective mechanisms in response to higher levels of CAS, it is relevant to note that teleost cytotoxic cells appear to have complex pathways of regulation of activation-induced cell death. Previous studies have demonstrated the inability of NCC to recycle in in vitro assays, similar to 237 what has been described for mammalian NK cells [27]. We propose that a mechanism of tight regulation of cell death in response to adverse conditions, as in the case of SASF or TNF alpha secretion, could be an effective substitute for recycling of effector cells to maintain a heightened state of innate immunity. It is important to identify other key players involved in these complex pathways in order to design effective prophylactic therapies. ACKNOWLEDGEMENTS This research was funded in part by grants from BARD US-3159-99C. REFERENCES [1] Xiao, Z., McGrew, J.T., Schroeder, A.J. & Fitzgerald-Hayes, M. (1993). CSE1 and CSE2, two new genes required for accurate mitotic chromosome segregation in Saccharomyces cerevisiae. Molecular & Cellular Biology 13, 4691-4702. [2] Brinkmann, U., Brinkmann, E., Gallo, M. & Pastan, I. (1995). Cloning and Characterization of A Cellular Apoptosis Susceptibility Gene, the Human Homolog to the Yeast Chromosome Segregation Gene Cse1. Proceedings of the National Academy of Sciences of the United States of America 92, 10427-10431. [3] Brinkmann, U., Brinkmann, E. & Pastan, I. (1995). Expression cloning of cDNAs that render cancer cells resistant to Pseudomonas and diphtheria toxin and immunotoxins. Molecular Medicine 1, 206-216. [4] Brinkmann, U., Gallo, M., Polymeropoulos, M.H. & Pastan, I. (1996). The human CAS (cellular apoptosis susceptibility) gene mapping on chromosome 20q13 is amplified in BT474 breast cancer cells and part of aberrant chromosomes in breast and colon cancer cell lines. Genome Research 6, 187-194. [5] Brinkmann, U. (1998). CAS, the human homologue of the yeast chromosome-segregation gene CSE1, in proliferation, apoptosis, and cancer. American Journal of Human Genetics 62, 509-513. [6] Behrens, P., Brinkmann, U. & Wellmann, A. (2003). CSE1L/CAS: its role in proliferation and apoptosis. Apoptosis 8, 39-44. 238 [7] Brinkmann, U., Brinkmann, E., Gallo, M., Scherf, U. & Pastan, I. (1996). Role of CAS, a human homologue to the yeast chromosome segregation gene CSE1, in toxin and tumor necrosis factor mediated apoptosis. Biochemistry 35, 6891-6899. [8] Scherf, U., Kalab, P., Dasso, M., Pastan, I. & Brinkmann, U. (1998). The hCSE1/CAS protein is phosphorylated by HeLa extracts and MEK-1: MEK-1 phosphorylation may modulate the intracellular localization of CAS. Biochemical & Biophysical Research Communications 250, 623-628. doi:10.1006/bbrc.1998.9367 [9] Wellmann, A., Krenacs, L., Fest, T., Scherf, U., Pastan, I., Raffeld, M. & Brinkmann, U. (1997). Localization of the cell proliferation and apoptosis-associated CAS protein in lymphoid neoplasms. American Journal of Pathology 150, 25-30. [10] Wellmann, A., Flemming, P., Behrens, P., Wuppermann, K., Lang, H., Oldhafer, K., Pastan, I. & Brinkmann, U. (2001). High expression of the proliferation and apoptosis associated CSE1L/CAS gene in hepatitis and liver neoplasms: correlation with tumor progression. International Journal of Molecular Medicine 7, 489-494. [11] Boni, R., Wellmann, A., Man, Y.G., Hofbauer, G. & Brinkmann, U. (1999). Expression of the proliferation and apoptosis-associated CAS protein in benign and malignant cutaneous melanocytic lesions. American Journal of Dermatopathology 21, 125-128. [12] Behrens, P., Brinkmann, U., Fogt, F., Wernert, N. & Wellmann, A. (2001). Implication of the proliferation and apoptosis associated CSE1L/CAS gene for breast cancer development. Anticancer Research 21, 2413-2417. [13] Gorlich, D., Dabrowski, M., Bischoff, F.R., Kutay, U., Bork, P., Hartmann, E., Prehn, S. & Izaurralde, E. (1997). A novel class of RanGTP binding proteins. Journal of Cell Biology 138, 65-80. [14] Fornerod, M., van Deursen, J., van Baal, S., Reynolds, A., Davis, D., Murti, K.G., Fransen, J. & Grosveld, G. (1997). The human homologue of yeast CRM1 is in a dynamic subcomplex with CAN/Nup214 and a novel nuclear pore component Nup88. EMBO Journal 16, 807-816. [15] Kutay, U., Bischoff, F.R., Kostka, S., Kraft, R. & Gorlich, D. (1997). Export of importin alpha from the nucleus is mediated by a specific nuclear transport factor. Cell 90, 10611071. doi:10.1016/S0092-8674(00)80372-4 [16] Graves, S.S., Evans, D.L. & Dawe, D.L. (1985). Antiprotozoan activity of nonspecific cytotoxic cells (NCC) from the channel catfish (Ictalurus punctatus). Jounal of Immunology 134, 78-85. [17] Evans, D.L., Jaso-Friedmann, L., Smith, E.E., St John, A., Koren, H.S. & Harris, D.T. (1988). Identification of a putative antigen receptor on fish nonspecific cytotoxic cells with monoclonal antibodies. Journal of Immunol 141, 324-332. 239 [18] Jaso-Friedmann, L., Harris, D.T., St John, A., Koren, H.S. & Evans, D.L. (1990). A monoclonal antibody-purified soluble target cell antigen inhibits nonspecific cytotoxic cell activity. Journal of Immunology 144, 2413-2418. [19] Evans, D.L., Leary, J.H. & Jaso-Friedmann, L. (1998). Nonspecific cytotoxic cell receptor protein-1: a novel (predicted) type III membrane receptor on the teleost equivalent of natural killer cells recognizes conventional antigen. Cellular Immunology 187, 19-26. doi:10.1006/cimm.1998.1310 [20] Faisal. M., Ahmed, I.I., Peters, G. & Cooper E.L. (1989). Natural Cytotoxicity of Tilapia Leukocytes. Diseases of Aquatic Organisms 7, 17-22. [21] Greenlee, A.R., Brown, R.A. & Ristow, S.S. (1991). Nonspecific cytotoxic cells of rainbow trout (Oncorhynchus mykiss) kill YAC-1 targets by both necrotic and apoptic mechanisms. Developmental & Comparative Immunology 15, 153-164. doi:10.1016/0145-305X(91)90006-K [22] McKinney, E.C. & Schmale, M.C. (1994). Damselfish with neurofibromatosis exhibit cytotoxicity toward tumor targets. Developmental & Comparative Immunology 18, 305313. doi:10.1016/S0145-305X(94)90356-5 [23] Suzumura, E., Kurata, O., Okamoto, N. & Ikeda, Y. (1994). Characteristics of Natural Killer-Like Cells in Carp. Fish Pathology 29, 199-203. [24] Jaso-Friedmann, L. & Evans, D.L. (1999). Mechanisms of cellular cytotoxic innate resistance in tilapia (Oreochromis nilotica). Developmental & Comparative Immunology 23, 27-35. doi:10.1016/S0145-305X(98)00047-0 [25] Jaso-Friedmann, L., Peterson, D.S., Gonzalez, D.S. & Evans, D.L. (2002). The antigen receptor (NCCRP-1) on catfish and zebrafish nonspecific cytotoxic cells belongs to a new gene family characterized by an F-box-associated domain. Journal of Molecular Evolution 54, 386-395. [26] Ullberg, M. & Jondal, M. (1981) Recycling and target binding capacity of human natural killer cells. Journal of Experimental Medicine 153, 615-628. [27] Evans, D.L., Carlson, R.L., Graves, S.S. & Hogan, K.T. (1984). Nonspecific cytotoxic cells in fish (Ictalurus punctatus). IV. Target cell binding and recycling capacity. Developmental & Comparative Immunology 8, 823-833. doi:10.1016/0145305X(84)90065-X [28] Bishop, G.R., Jaso-Friedmann, L. & Evans, D.L. (2000). Activation-induced programmed cell death of nonspecific cytotoxic cells and inhibition by apoptosis regulatory factors. Cellular Immunology 199, 126-137. doi:10.1006/cimm.1999.1609 [29] Evans, D.L., Leary, J.H. & Jaso-Friedmann, L. (2001). Nonspecific cytotoxic cells and innate immunity: regulation by programmed cell death. Developmental & Comparative Immunology 25, 791-805. doi:10.1016/S0145-305X(01)00036-2 240 [30] Jaso-Friedmann, L., Ruiz, J., Bishop, G.R. & Evans, D.L. (2000). Regulation of innate immunity in tilapia: activation of nonspecific cytotoxic cells by cytokine-like factors. Developmental & Comparative Immunology 24, 25-36. doi:10.1016/S0145305X(99)00053-1 [31] Evans, D.L., Taylor, S.L., Leary, J.H. Bishop, G.R., Eldar, A. & Jaso-Friedmann, L. (2000). In vivo activation of tilapia nonspecific cytotoxic cells by Streptococcus iniae and amplification with apoptosis regulatory factor(s). Fish & Shellfish Immunology 10, 419434. doi:10.1006/fsim.1999.0250 [32] Marchler-Bauer, A., Anderson, J.B., Cherukuri, P.F., Weese-Scott, C., Geer, L.Y., Gwadz, M., He, S., Hurwitz, D.I., Jackson, J.D., Ke, Z., Lanczycki, C.J., Liebert, C.A., Liu, C., Lu, F., Marchler, G.H., Mullokandov, M., Shoemaker, B.A., Simonyan, V., Song, J.S., Thiessen, P.A., Yamashita, R.A., Yin, J.J., Zhang, D. & Bryant, S.H. (2005). CDD: a Conserved Domain Database for protein classification. Nucleic Acids Research 33, D192-D196. [33] Praveen, K., Evans, D.L. & Jaso-Friedmann, L. (2004). Evidence for the existence of granzyme-like serine proteases in teleost cytotoxic cells. Journal of Molecular Evolution 58, 449-459. [34] Heaton, M.P., LopezCorrales, N.L., Smith, T.P.L., Kappes, S.M. & Beattie, C.W. (1997). Directed cosmid isolation of bovine markers for physical assignment by fish. Animal Biotechnology 8, 167-177. [35] Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, Z., Miller, W. & Lipman, D.J. (1997). Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Research 25, 3389-3402. [36] Kumar, S., Tamura, K., Jakobsen, I.B. & Nei, M. (2001). MEGA2: molecular evolutionary genetics analysis software. Bioinformatics 17, 1244-1245. 241 Figure 7.1. Compiled full-length cDNA sequence of tilapia CAS. The start and stop codons are represented in bold. Polyadenylation signal is highlighted and poly-A tail is underlined. The highlighted portion in the amino acid chain represents the highly conserved Zn finger domain. Construction of cDNA libraries from NCC of different fish species has been described previously [33]. A directed PCR-based iterative screening protocol was used to identify positive clones for CAS [34]. Specific primers were designed based on conserved regions in various CAS sequences were identified by multiple sequence alignments using Clustal W provided with vector NTI package, version 6 (InforMax Inc, Carlsbad, CA). Sense (5’-cagtctgtgaaaggccacactataag-3’) and antisense (5’-tcattggcttgtgttattccatgcttctg-3’) PCR primers were used to amplify a portion of tilapia CAS gene using tilapia NCC cDNA and bacterial pellet of tilapia NCC cDNA library as a template. The amplicons were TA cloned in to pDrive cloning vector using a PCR cloning kit (Qiagen, Valencia, CA) and sequenced to verify the identity. After several rounds of screening, the culture-enriched for target DNA was spread on solid medium and colonies were randomly selected for assay by PCR. Several CAS positive clones were isolated, expanded and frozen in glycerol stocks for further analysis. These clones were sequenced in a 373 A DNA sequencer (Applied Biosystems, CA). The full length tilapia CAS cDNA sequence was obtained by doing 5’ and 3’ RACE on fresh mRNA isolated from tilapia NCC. The entire sequence was compared with the known sequences in DDBJ/EMBL/GenBank databases using BLAST version 2.2.5 [35]. 242 1 51 101 151 201 251 301 351 401 451 501 551 601 651 701 751 801 851 901 951 1001 1051 1101 1151 1201 1251 1301 GCATTTGAAAGCTGCCCTGGCTCGGGAAAGCGGCACGTTCTGTACGAACT TGTGGTGGCTCTCCATCCCATCCCTAAATTAAACTCACTATCACGTTTTC M E L N D A N L Q T L T E F L R ATCATGGAGCTGAATGATGCTAACCTACAGACCCTGACTGAGTTCCTTAG K T L D P D P T V R R P A E K F AAAAACCCTCGACCCAGATCCAACAGTCAGACGCCCTGCTGAAAAGTTTC L E S V E G N Q N Y P L L L L T L TTGAATCTGTAGAAGGAAACCAGAACTATCCACTGTTACTTCTCACGTTG L E K S Q D N V I R V C A A V T F CTGGAGAAGTCCCAGGACAACGTGATCCGTGTCTGTGCTGCTGTGACATT K N Y I K R N W R I V E D E P N CAAAAACTATATAAAAAGAAACTGGAGAATTGTTGAAGATGAACCCAACA K I S D P D R T A V K A N I V N L AAATCTCTGATCCTGACCGAACGGCAGTAAAAGCAAACATTGTAAATTTG M L S S P E Q I Q K Q L S D A I S ATGCTGAGCAGCCCAGAACAGATTCAGAAACAGTTAAGTGATGCCATCAG I I G R E D F P Q K W P D L L T CATCATAGGACGTGAAGACTTTCCACAAAAATGGCCAGACCTTCTAACAG E M V A R F R S G D F H I I N G V AGATGGTGGCTCGCTTCAGAAGTGGAGATTTCCACATCATCAATGGTGTG L R T A H S L F K R Y R H E F K S CTGCGCACTGCACATTCGCTCTTCAAGAGGTATCGCCATGAGTTCAAGTC N E L W S E I K L V L D T F A L AAATGAGCTTTGGTCTGAGATAAAACTGGTTCTGGACACGTTTGCTCTAC P L T E L F K A T I E L C Q T H A CTCTGACTGAACTATTCAAGGCCACAATCGAGCTGTGTCAAACTCATGCT T D V N A L K V L F S S L T L I A ACAGATGTCAATGCCTTGAAGGTCCTCTTTTCATCACTCACACTCATCGC K L F Y S L N F Q D L P E F F E CAAGCTTTTCTACAGTCTTAACTTTCAGGACCTTCCAGAGTTTTTTGAAG D N M E T W M T N F H G L L T L D ACAATATGGAAACCTGGATGACCAATTTCCACGGCCTACTGACTTTGGAT N K L L Q T D D E E E A G L L E L AATAAACTTTTACAAACAGATGATGAGGAGGAGGCAGGTCTCCTGGAGCT L K S Q I C D N A A L Y A Q K Y GCTGAAGTCTCAGATCTGTGACAATGCGGCTCTTTATGCTCAGAAGTACG D E E F Q P Y L P R F V T A I W N ATGAGGAATTTCAGCCATACCTGCCGCGCTTTGTTACTGCAATCTGGAAC L L V S T G Q E V K Y D L L V S N CTTCTAGTTTCTACTGGCCAGGAAGTCAAATATGACTTGCTCGTAAGCAA A I Q F L A S V C E R P H Y K H TGCTATCCAGTTCTTGGCATCAGTCTGTGAAAGGCCACACTATAAGCATT L F E D Q N T L T S I C E K V I V TGTTTGAGGACCAGAATACACTCACAAGCATTTGTGAGAAGGTCATTGTG P N M E F R S A D E E A F E D N S CCCAACATGGAGTTCAGAAGTGCGGATGAGGAGGCCTTTGAAGATAACTC E E Y I R R D L E G S D I D T R GGAGGAGTACATCCGAAGAGACCTGGAGGGATCTGACATTGACACTCGCC R R A A C D L V R G L C K F F E G GTAGGGCTGCATGTGATTTGGTGAGAGGGCTTTGTAAATTTTTTGAGGGA P V T A I F S G Y V N S M L S E Y CCAGTCACAGCAATCTTCTCCGGCTACGTGAACTCGATGCTGAGCGAGTA 243 16 32 49 66 82 99 116 132 149 166 182 199 216 232 249 266 282 299 316 332 349 366 382 399 416 1351 1401 1451 1501 1551 1601 1651 1701 1751 1801 1851 1901 1951 2001 2051 2101 2151 2201 2251 2301 2351 2401 2451 2501 2551 2601 A K N P R E N W K H K D A A I Y TGCCAAGAACCCTCGGGAGAACTGGAAACACAAGGATGCTGCCATCTATT L V T S L A S K A Q T Q K H G I T TGGTCACATCGCTGGCATCGAAAGCCCAGACACAAAAGCATGGAATAACA Q A N E L V N L T E F F V N H I L CAAGCCAACGAGTTGGTGAATCTGACAGAGTTCTTTGTGAACCACATTCT P D L K S P N V N E F P V L K A CCCTGATTTAAAATCCCCCAATGTTAATGAGTTCCCAGTGCTGAAGGCTG D A I K Y V M I F R S Q L P K E Q ATGCCATCAAGTATGTAATGATCTTCAGAAGTCAGCTTCCTAAAGAGCAG L L Q A V P L L I T H L Q A E S T CTGCTGCAGGCAGTCCCTCTACTAATAACTCACCTGCAGGCAGAGAGCAC V E H T Y A A H A L E R L F T M AGTGGAGCACACTTATGCTGCACATGCTTTGGAGAGACTGTTCACTATGA R G P N N A T L I T A A E M A P F GGGGACCCAACAACGCAACACTTATCACTGCTGCAGAGATGGCACCGTTT T E Q L L N N L F K A L A F P G S ACTGAACAGCTGCTCAACAACTTGTTCAAGGCACTGGCTTTTCCTGGTTC A E N E Y I M K A I M R S F S L TGCAGAAAATGAATACATCATGAAAGCCATCATGCGCAGCTTCTCCCTGC L Q E S I V P Y I P T L I G Q L T TGCAGGAGTCAATTGTTCCCTACATCCCAACACTGATTGGTCAGCTCACT H K L L Q V S K N P S K P H F N H CATAAGCTCCTCCAAGTCAGCAAGAATCCCAGCAAACCTCACTTTAACCA Y L F E S L C L S V R I T C K A CTACCTGTTTGAGTCCCTGTGCCTGTCTGTCCGCATCACCTGCAAGGCAA N P T T V S S F E E A L F P V F T ACCCCACTACTGTCAGCAGTTTCGAGGAAGCACTCTTCCCTGTCTTCACA E I L Q N D V Q E F L P Y V F Q V GAGATCCTTCAGAACGATGTTCAGGAGTTTCTTCCATATGTGTTCCAGGT M S L L L E I H S N S I P A S Y GATGTCTCTCCTCCTAGAGATCCACTCCAACTCTATTCCCGCTTCCTATA M A L F P H L L Q P V L W E R T G TGGCTTTATTCCCTCACCTGCTGCAGCCTGTACTTTGGGAACGAACAGGG N I P P L V R L L Q A Y L E K G G AACATCCCCCCTCTGGTGCGCCTCCTTCAGGCTTACCTGGAGAAGGGAGG E T I A R S A A D K I P G L L G TGAAACTATTGCTAGGTCTGCTGCTGATAAAATACCTGGCTTGCTTGGAG V F Q K L I A S K A N D H Q G F Y TTTTCCAAAAGCTTATAGCATCCAAGGCCAATGATCATCAAGGATTTTAC L L N S I I E H M P P E S L T Q Y CTTCTCAACAGCATCATAGAGCACATGCCCCCAGAATCACTCACTCAGTA R K Q I F I L L F Q R L Q S S K CAGGAAACAGATCTTCATTTTGCTCTTCCAGAGGCTACAAAGTTCCAAAA T T K F I K S F L V F V N L Y C V CCACCAAGTTCATCAAGAGTTTCCTGGTGTTTGTCAATTTGTATTGTGTC K Y G A I A L Q E I F D S I Q P K AAATATGGAGCTATAGCACTTCAGGAGATTTTTGACAGCATCCAGCCAAA M F G M V L E K I V I P E V Q K AATGTTTGGTATGGTGCTGGAGAAAATAGTTATTCCAGAGGTTCAGAAGG V S G P V E K K I C A V G I T K V TGTCTGGACCAGTCGAGAAGAAGATCTGTGCTGTCGGCATTACAAAGGTC 244 432 449 466 482 499 516 532 549 566 582 599 616 632 649 666 682 699 716 732 749 766 782 799 816 832 849 2651 2701 2751 2801 2851 2901 2951 3001 3051 3101 3151 3201 3251 3301 3351 3401 3451 3501 3551 L T E C P A M M D T E Y T K L W T CTCACTGAGTGTCCTGCAATGATGGACACGGAGTACACGAAACTCTGGAC P L L Q A L I G L F E L P E D D CCCACTGCTCCAGGCCCTCATTGGTTTATTTGAGCTACCTGAAGATGACA S I P D D E H F I D I E D T P G Y GCATCCCAGACGACGAGCACTTCATTGACATCGAAGACACACCGGGCTAT Q T A F S Q L A F A G K K E H D P CAGACCGCCTTCTCACAGCTGGCCTTTGCTGGCAAGAAGGAGCACGACCC I G D A V G N P K I L L A Q S L AATTGGAGATGCTGTTGGCAATCCAAAAATTCTGCTGGCCCAGTCACTGC H K L S T A C P G R V P S M L S T ACAAGCTTTCTACTGCCTGTCCTGGAAGGGTTCCTTCAATGCTGAGCACG S L N A E A L Q F L Q G Y L Q A A AGTCTGAATGCAGAAGCTCTCCAGTTCCTGCAGGGGTACTTGCAGGCGGC T V Q L V * TACTGTGCAGCTGGTTTAAAACCAAAGCGAGGGTGTTCACACAAAGCAAA ATCTCAGTACACTGCAGGAATAGCCCCCAATGGCCCCCCAGTCACATGAC AGTCCTGCTTCAGTTGAACTCGAGACCTGAAAGTTAAATGATGGATTTAC CAGAGAAGTAAATTTTGCCTGAATTCAGGGTGTGTACTCCAGTGGAAGTC ATTTTAAGGTTTTTCTTTTAAAGAAAAAAGGAACACAACCAAAATGTTAT TACTTTAAGATATCTGGGCCTTTTTATTTCCTGTGTTAAAGGCTAACAAG GATTTGGATGTCAGGTTGTGAAGCACTTGCCGGTTTATTTCAAAAACATC AGCAATAGAAGCAGATGTAAAAAACTGAACATGTGGGACACTGAACATCT TTCAAGCACCGCATCCGCAAAGAGGAGTCCATGCGCTGGTTCCAGCAGAA ATACGACGGCATCATCCTCCCCGGCAAGTAAAGGACCACCTTTTGCTCTT GTTTGTAACAAAGTAATAAAATGGAAAAAATAAAAAAAAAAAAAAAAAAA AAAAAAAAAAAAAAAAAAAAAA 245 866 882 899 916 932 949 966 971 Figure 7.2. Multiple sequence alignment of tilapia CAS with similar proteins from other species. Multiple sequence alignments were done with Clustal W using Vector NTi suite version 6 (InforMax Inc). 246 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : MELNDANLQTLTEFLRKTLDPDPTVRRPAEKFLESVEGNQNYPLLLLTLLEKSQDNVIRVCAAVTFKNYIK MELNDANLQTLTEFLRKALDPDPTVRRPAEKFLESVEGNQNYPLLLLTLLEKSQDNVIRVCAAVTFKNYIK MELNDGNLQTLTEYLQKTLSADPAVRRPAEKFLESVEGNQNYPILLLTVLEKSQNEVIRVCSAVTFKNYIK MELSEGNLQGLTEYLKKTLDPDPAVRRPAEKYLESVEGNQNYPLLLLTLVERSQDNVIKVCSAVTFKNYIK MELSDANLQTLTEYLKKTLDPDPAIRRPAEKFLESVEGNQNYPLLLLTLLEKSQDNVIKVCASVTFKNYIK MELSDANLQTLTEYLKKTLDPDPAIRRPAEKFLESVEGNQNYPLLLLTLLEKSQDNVIKVCASVTFKNYIK MELSDANLQTLTEYLKKTLDPDPAIRRPAEKFLESVEGNQNYPLLLLTLLEKSQDNVIKVCASVTFKNYIK : : : : : : : 71 71 71 71 71 71 71 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : RNWRIVEDEPNKISDPDRTAVKANIVNLMLSSPEQIQKQLSDAISIIGREDFPQKWPDLLTEMVARFRSGD RNWRVIEDEPNKVSDPDRTAIKANIVNLMLSSPEQIQKQLSDAISIIGREDFPQKWPDLLTEMVTRFRSGD RNWRIVEDEPNKISDPDRTAIKANIVNLMLTSPEQIQKQLSDAISIIGREDFPLKWPDLLTEMVNRFQSGD RNWRIVEDESNKICEADRIAVKSSIINLMLRSPEQIQKQLSDAISIIGREDFPQKWPNLLTEMVNRFQSGD RNWRIVEDEPNKICEADRVAIKANIVHLMLSSPEQIQKQLSDAISIVGREDFPQKWPDLLTEMVNRFQSGD RNWRIVEDEPNKICEADRVAIKANIVHLMLSSPEQIQKQLSDAISIIGREDFPQKWPDLLTEMVNRFQSGD RNWRIVEDEPNKICEADRVAIKANIVHLMLSSPEQIQKQLSDAISIIGREDFPQKWPDLLTEMVNRFQSGD : : : : : : : 142 142 142 142 142 142 142 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : FHIINGVLRTAHSLFKRYRHEFKSNELWSEIKLVLDTFALPLTELFKATIELCQTHATDVNALKVLFSSLT FHIINGVLRTAHSLFKRYRHEFKSNELWSEIKLVLDTFALPLTELFKATIELCQTHATDVNALKVLFSSLT FHIINGVLRTAHSLFKRYRHEFKSNELWSEIKLVLDTFAQPLTELFKATIELCQTHATDINALKVLFSSLT FHVINGVLHTAHSLFKRYRHEFKSSELWTEIKLVLDTFAGPLTDLFKATIELCNTHANDVGALKVLFSSLN FHVINGVLRTAHSLFKRYRHEFKSNELWTEIKLVLDAFALPLTNLFKATIELCSTHANDASALRILFSSLI FHVINGVLRTAHSLFKRYRHEFKSNELWTEIKLVLDAFALPLTNLFKATIELCSTHANDASALRILFSSLI FHVINGVLRTAHSLFKRYRHEFKSNELWTEIKLVLDAFALPLTNLFKATIELCSTHANDASALRILFSSLI : : : : : : : 213 213 213 213 213 213 213 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : LIAKLFYSLNFQDLPEFFEDNMETWMTNFHGLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LISKLFYSLNFQDLPEFFEDNMETWMTNFHGLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LISKLFYSLNFQDLPEFFEDNMETWMTNFHNLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LIAKLFHSLNFQDLPEFFEDNMETWMTNFHNLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LISKLFYSLNFQDLPEFFEDNMETWMNNFHTLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LISKLFYSLNFQDLPEFFEDNMETWMNNFHTLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE LISKLFYSLNFQDLPEFWEGNMETWMNNFHTLLTLDNKLLQTDDEEEAGLLELLKSQICDNAALYAQKYDE : : : : : : : 284 284 284 284 284 284 284 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : EFQPYLPRFVTAIWNLLVSTGQEVKYDLLVSNAIQFLASVCERPHYKHLFEDQNTLTSICEKVIVPNMEFR EFQPYLPRFVTAIWNLLVSTGQEVKYDLLVSNAIQFLASVCERPHYKHLFEDQNTLTSICEKVIVPNMEFR EFQPYLPRFVTAIWNLLVTTGQEVKYDLLVSNAIQFLASVCERPHYKHLFEDQNVLTSICEKVIVPNMEFR EFQPYLPRFVTAIWNLLVTTGQEVKYDLLVSNAIQFLASVCERPHYKNLFEDPSTLTSICEKVIVPNMEFR EFQRYLPRFVTAIWNLLVTTGQEVKYDLLVSNAIQFLASVCERPHYKNLFEDQNTLTSICEKVIVPNMEFR EFQRYLPRFVTAIWNLLVTTGREVKYDLLVSNAIQFLASVCERPHYKNLFEDQNTLTSICEKVIVPNMEFR EFQRYLPRFVTAIWNLLVTTGQEVKYDLLVSNAIQFLASVCERPHYKNLFEDQNTLTSICEKVIVPNMEFR : : : : : : : 355 355 355 355 355 355 355 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : SADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTAIFSGYVNSMLSEYAKNPRENWKH SADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTAIFSGYVNSMLAEYAKNPGENWKH SADEEAFEDNSEEYIIRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTGIFSGYVNSMLAEYAKNPGVNWKH AADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTNIFSGYVNSMLQEYAKNPSVNWKH AADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTGIFSGYVNSMLQEYAKNPSVNWKH AADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTGIFSGYVNSMLQEYAKNPSVNWKH AADEEAFEDNSEEYIRRDLEGSDIDTRRRAACDLVRGLCKFFEGPVTGIFSGYVNSMLQEYAKNPSVNWKH : : : : : : : 426 426 426 426 426 426 426 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : KDAAIYLVTSLASKAQTQKHGITQANELVNLTEFFVNHILPDLKSPNVNEFPVLKADAIKYVMIFRSQLPK KDAAIYLVTSLASKAQTQKHGITQANELVNLNEFFVNHILSDLKSHNVNEFPVLKADAIKYVMIFRSQLPK KDAAIYLVTSLASKAQTQKHGITQANELVNLSEFFLNHILIDLKSPNVNEFPVLKSDAIKYVMTFRSQLPK KDAAIYLVTSLASKAQTQKHGITQANELVNLTEFFVNHILPDLKSANINQYPVLKADGIKYIMFFRSQIPR KDAAIYLVTSLASKAQTQKHGITQANELVNLTEFFVNHILPDLKSNNVNEFPVLKADGIKYIMIFRNQVPK KDAAIYLVTSLASKAQTQKHGITQANELVNLTEFFVNHILPDLKSNNVNEFPVLKADGIKYIMIFRNQVPK KDAAIYLVTSLASKAQTQKHGITQANELVNLTEFFVNHILPDLKSANVNEFPVLKADGIKYIMIFRNQVPK : : : : : : : 497 497 497 497 497 497 497 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : EQLLQAVPLLITHLQAESTVEHTYAAHALERLFTMRGPNNATLITAAEMAPFTEQLLNNLFKALAFPGSAE EQLLQAVPLLISHLQAESTVEHTYAAHALERLFTMRGPNNTTLITPVEMAPFTEQLLNNLFKSLALPGSAE EQLLQAVPLLVSHLQAESIVQHTYAAHALERLFTMRGGNNTTLITPTEMAPFTEQLLNHLFKALAIPGSSE EQLLVTIPLLIAYLQAESIVVHTYAAHALERFFTMKGAATTTLIVAADMMPYVELLLANLFKALSLPGSTE EHLLVSIPLLISHLGAESIVVHTYAAHALERLFTMRGPNNATLFTAAEIAPFVGILLTNLFKALTLPGSSE EHLLVSIPLLISHLEAESIVVHTYAAHALERLFTMRGSNNTTLFTAAEIAPFVEILLTNLFKALTLPGSSE EHLLVSIPLLINHLQAGSIVVHTYAAHALERLFTMRGPNNATLFTAAEIAPFVEILLTNLFKALTLPGSSE : : : : : : : 568 568 568 568 568 568 568 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : NEYIMKAIMRSFSLLQESIVPYIPTLIGQLTHKLLQVSKNPSKPHFNHYLFESLCLSVRITCKANPTTVSS NEYIMKAIMRTFSLLQEAIVPYIPTLIGQLTHKLLLVSKNPSKPHFNHYLFESLCLSVRITCKANPATVSS NEYIMKAIMRSFSLLQEAIVPYIPTLIGQLTHKLLLVSKNPSKPHFNHYLFESLCLSIRITCKANPDTVSS NEYIMKAIMRSFSLLQEAIIPYIPSVISQLTQKLLAVSKNPSKPHFNHYMFEAICLSIRITCRANPAAVAS NEYIMKAIMRSFSLLQEAIIPYIPTLITQLTQKLLAVSKNPSKPHFNHYMFEAICLSIRITCKANPAAVVN NEYIMKAIMRSFSLLQEAIIPYIPTLITQLTQKLLAVSKNPSKPHFNHYMFEAICLSIRITCKANPAAVVN NEYIMKAIMRSFSLLQEAIIPYIPTLITQLTQKLLAVSKNPSKPHFNHYMFEAICLSIRITCKANPAAVVN : : : : : : : 639 639 639 639 639 639 639 247 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : FEEALFPVFTEILQNDVQEFLPYVFQVMSLLLEIHSNSIPASYMALFPHLLQPVLWERTGNIPPLVRLLQA FEEALFPVFTEILQNDVQEFLPYVFQVMSLLLEIHSSSIPSSYMALFPHLLQPALWERTGNIPPLVRLLQA FEEALFPVFTEILQNDVQEFVPYVFQVMSLLLEIHSNSIPSSYMALFPHLLQPVLWERTGNIPPLVRLLQA FEDALFLVFTEILQSDVQEFIPYVFQVMSLLLEIHTTDIPPSYMALFPHLLQPVLWERTGNIPPLVRLLQA FEEALFLVFTEILQNDVQEFIPYVFQVMSLLLETHKNDIPSSYMALFPHLLQPVLWERAGNIPALVRLLQA FEEALFLVFTEILQNDVQEFIPYVFQVMSLLLETHKNDIPSSYMALFPHLLQPVLWERTGNIPALVRLLQA FEEALFLVFTEILQNDVQEFIPYVFQVMSLLLETHKNDIPSSYMALFPHLLQPVLWERTGNIPALVRLLQA : : : : : : : 710 710 710 710 710 710 710 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : YLEKGGETIARSAADKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHMPPESLTQYRKQIFILLFQRLQSS YLEKGGATIAASAADKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHMPPESITQYRKQIFILLFQRLQSS YLEKGAAAIANTASDKIPGLLGVFQKLIASKANDHQGFYLLNSIVEHMPAEAITQYRKQIFILLFQRLQSS YLERGATTIAASASDKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHLPAECIEQYKRQIFIVLFQRLQSS FLERGSSTIATAAADKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHMPPESVDQYRKQIFILLFQRLQNS FLERGSSTIATAAADKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHMPPESVDQYRKQIFILLFQRLQNS FLERGSNTIASAAADKIPGLLGVFQKLIASKANDHQGFYLLNSIIEHMPPESVDQYRKQIFILLFQRLQNS : : : : : : : 781 781 781 781 781 781 781 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : KTTKFIKSFLVFVNLYCVKYGAIALQEIFDSIQPKMFGMVLEKIVIPEVQKVSGPVEKKICAVGITKVLTE KTTKFIKSFLVFVNLYSVKYGAIALQEIFDSIQPKMFGMVLEKIIIPEVQKVSGAVEKKICAVGITKVLTE KTTKFVKSFLVFINLYSVKYGAIALQEIFDDIQPKMFGMVVEKIVIPEVQKVSGQVEKKICAVGIIKILTE KTTKFVKSFLVFLNLFCIKFGAIALQEMFDSIQPKMFGMVVEKIIIPEIQKVSGPIEKKICAVGLTKVLTE KTTKFIKSFLVFINLYCIKYGALALQEIFDGIQPKMFGMVLEKIIIPEIQKVSGNVEKKICAVGITKLLTE KTTKFIKSFLVFINLYCIKYGALALQEIFDGIQPKMFGMVLEKIIIPEIQKVSGNVEKKICAVGITKLLTE KTTKFIKSFLVFINLYCIKYGALALQEIFDGIQPKMFGMVLEKIIIPEIQKVSGNVEKKICAVGITNLLTE : : : : : : : 852 852 852 852 852 852 852 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : CPAMMDTEYTKLWTPLLQALIGLFELPEDDSIPDDEHFIDIEDTPGYQTAFSQLAFAGKKEHDPIGDAVGN CPAMMDTEYTKLWTPLLQALIGLFELPEDDSIPDDEHFIDIEDTPGYQTAFSQLAFAGKKEHDPIGDAVGN CPAMMDTEYTKLWAPLLQALIGLFELPEDDSIPDDEHFIDIEDTPGYQTAFSQLAFAGKKEHDPIGDAVSN CPVMMDTEYTKLWTPLLQALIGLFELPEDDTIPDDEHFIDIEDTPGYQAAFSQLAFAGKKEHDPIGEMVNN CPAMMDTEYTKLWTPLLQSLIGLFELPEDDSIPDEEHFIDIEDTPGYQTAFSQLAFAGKKEYDPVGQTVNN CPPMMDTEYTKLWTPLLQSLIGLFELPEDDSIPDEEHFIDIEDTPGYQTAFSQLAFAGKKEHDPVGQMVNN CPPMMDTEYTKLWTPLLQSLIGLFELPEDDTIPDEEHFIDIEDTPGYQTAFSQLAFAGKKEHDPVGQMVNN : : : : : : : 923 923 923 923 923 923 923 Tilapia CAS AAN52370 Pagrus CAS BAA89430 Danio CAS AAH52479 Xenopus CAS AAH73735 Rat XP_342582 Mouse CAS AAG24636 Human CAS AAC50367 : : : : : : : PKILLAQSLHKLSTACPGRVPSMLSTSLNAEALQFLQGYLQAATVQLV PKILLAQSLHKLSTACPGRVPSMLSTSLNAEALQFLQGYLQAATVQLV PKILLAQSLHKLSTACPGRVPSMLSTSLPTEALQFLQGYLQAATVQLV PKILLAQSLHKLSTACPGRVPSMISTSLNAEALQFLQGYLQAGSVSLV PRVHLAQALHRLSTACPGRVPSMVSTSLNAEALQYLQGYLQAASVTLL PKIHLAQSLHKLSTACPGRVPSMVSTSLNAEALQYLQGYLQAASVTLL PKIHLAQSLHMLSTACPGRVPSMVSTSLNAEALQYLQGYLQAASVTLL 248 : : : : : : : 971 971 971 971 971 971 971 Figure 7.3. Phylogenetic analysis of tilapia CAS. Similar analyses as those previously done with catfish granzyme and NCCRP-1 of zebrafish were performed [25,33]. Briefly, multiple sequence alignments were done using Clustal W, followed by phylogenetic analysis using Mega version 2.1 [36]. The aligned data set was analyzed by the criteria of maximum parsimony using the branch-and-bound algorithm. The reliability of the trees was tested using 1000 bootstrap replicates. The alignment was also analyzed by the neighbor joining method as implemented by Mega with 1000 bootstrap replications. For neighbor joining method, Poisson correction was used with the complete deletion option. 249 Rat XP 342582 100 Mouse CAS AAG24636 Human CAS AAC50367 100 Xenopus CAS AAH73735 Tetraodon CAS-like CAG06818 Danio CAS AAH52479 99 Pagrus CAS BAA89430 99 Tilapia CAS AAN52370 Drosophila CAS NP 523588 100 Anopheles CAS XP 311424 Arabidopsis CAS AAD20163 100 250 Yeast CSE O13671 C-elegans CAS NP 490716 Figure 7.4. Expression of tilapia CAS in different tissues. Total RNA was isolated from various tissues using RNeasy Mini Kit (Qiagen, CA) according to manufacturer’s recommendations. Synthesis of cDNA was done using a cDNA synthesis kit (Invitrogen, CA) using oligo-dT primer. RT-PCR analysis was performed with tilapia CAS specific primers using cDNA from various tissues. Lane 1, purified NCC; lane 2, total white blood cells; lane 3, kidney; lane 4, spleen; lane 5, red blood cells; lane 6, heart; lane 7, liver; lane 8, gill; lane 9, muscle. 251 252 Figure 7.6. Transcriptional regulation of tilapia CAS in NCC. Purified NCC were subjected to various treatments, total RNA was isolated and synthesis of cDNA was done using a cDNA synthesis kit (Invitrogen, CA) using olido-dT primer. PCR was done with initial 2 minutes denaturation at 94oC followed by 25 cycles of following cycle: denaturation at 94oC for 30 s, annealing at 57oC for 30 s, extension at 72oC for 30 s. The products were resolved on a 1.5% agarose gel and ethidium bromide-stained bands were quantified using a Kodak DC290 digital camera and digitized using UN-SCAN-IT software (Silk Scientific, UT). Band intensities (pixel values) were normalized to that of beta-actin. 253 A 0.5 0.45 0.4 CAS/Actin 0.35 0.3 0.25 0.2 0.15 0.1 0.05 0 Non-Stressed Stressed B 0.35 0.3 CAS/Actin 0.25 0.2 0.15 0.1 0.05 0 0H 2H 254 4H CHAPTER 8 SUMMARY AND CONCLUSIONS 255 Cell-mediated cytotoxicity is an important component of the immune system to destroy virally infected cells and transformed cells of a tumor. Cytotoxic lymphocytes are the main mediators of this function. These cells vary considerably with regard to the mechanisms they use to recognize the target cells. However, once the target is identified, effector cells use similar pathways to induce cellular death. The main pathways employed by these cells include the use of their cytotoxic granules and/or ligation of TNF superfamily members to corresponding death receptors on the target cells. The activation of these mechanisms leads to cell death though different cell death pathways. Even though the importance of these effector pathways in target cell death has been demonstrated, much remains to be understood regarding the molecular identity of effector molecules and their relative contribution to the cell death pathways. Novel components of cytotoxic granules are being described, while functions of many known components are still unknown. For example, multiple granzymes with different substrate specificities and unknown physiological functions are described from these killer cells. Furthermore, the roles of various TNF-superfamily members in different cytotoxic functions are still poorly understood. One of the most significant outcomes of understanding the effector molecules of cell-mediated cytotoxicity is that it will lead to the discovery of novel cell death and proliferation pathways. In addition, it is significant to understand the role of these molecules in the context of immune reactions, so that efficient anti-tumor and anti-viral strategies can be adopted to enhance the immune system during the course of an infection. Granzymes are members of the serine protease family expressed in the cytoplasmic granules of professional killer cells. Multiple granzymes with variable substrate specificities have been identified in cytotoxic lymphocytes. Most of the studies on granzymes were 256 concentrated on granzymes A and B, and evidence suggests very complicated pathways of cell death initiated by these proteases. Results from recent studies suggest that, in addition to A and B, other granzymes also play a significant role in inducing cell death pathways. Nonspecific cytotoxic cells are the first identified cytotoxic cell population in teleost and they are believed to be the evolutionary precursors of NK cells and CTLs. NCC can induce cell death in a variety of targets including virally infected cells, protozoan parasites and even mammalian tumor cells. This last characteristic is important because it appears to indicate that similar killing mechanisms may be used by cytotoxic cells of ectothermic and endothermic vertebrates. NCC do not require any prior activation and the killing is not MHC-restricted. Ease of purification of these cells and their cytotoxic effects on a variety of mammalian targets make them an ideal model system to study the cell-mediated cytotoxicity. Moreover, NCC play a significant role in the immune functions and in the health of teleosts. Previous studies have demonstrated various aspects of cytotoxicity mediated by NCC, including the interaction of NCC with the target cells. However, there was no molecular evidence that could be used to identify the effector molecules involved in the cytotoxic activity of NCC. In the present study, the identity of cytotoxic molecules involved in cell death pathways from two different species of fish is presented for the first time. The two cell death pathways initiated by NCC induce target cell death through granule exocytosis as well as TNF-superfamily ligands. Teleost cytotoxic cells express multiple granzymes and other components of the granule exocytosis pathway. Substrate specificities of individual teleost granzymes were assessed by expressing them as recombinant proteins and measuring the hydrolysis of thiobenzyl ester substrates. Teleost granzymes identified so far have tryptase and chymase activity, but no Asp- 257 ase or Met-ase activities. Tryptase, Asp-ase, Met-ase and chymase activities are the major substrate specificities associated with mammalian granzymes. Teleost cytotoxic cells exocytose the contents of their granules upon conjugate formation with the targets and cytotoxicity correlates with the residual granzyme activity in the supernatants. Catfish NCC constitutively express high level of granzymes, while tilapia NCC can be induced to upregulate the granzyme expression. Teleost granzymes do have a novel S1 specificity pocket triplet, which is not seen in any mammalian granzymes. Phylogenetic analysis of all known granzyme sequences suggests clustering of teleost chymases into a separate group. Teleost cytotoxic cells also express two forms (membrane-bound as well as secreted) of TNF-alpha. Both forms of TNF can induce cell death in susceptible targets. We have previously demonstrated the role of the secreted form of Fas ligand in inducing cell death by NCC. Here we show that TNF also functions as a cytokine by protecting the cells from activation-induced cell death and activating the NCC to express higher levels of granzymes. These results suggest a major role played by TNF superfamily members in NCC functions. Unlike mammalian NK cells, NCC lack the ability to recycle and kill multiple target cells during in vitro cytotoxicity assays. The importance of regulators of apoptosis in protecting NCC from activation-induced cell death during immune reactions has been proposed. This study demonstrates the upregulation of cellular apoptosis susceptibility (CAS) protein upon treatment with stress-activated serum factors and recombinant TNF-alpha, leading to protection of cells from AICD. It is proposed that CAS may be an important modulator of NCC functions. In conclusion, in this study, molecular evidence is presented for the first time for a parallel evolution of effector molecules of cell-mediated cytotoxicity in teleosts. These findings are significant because they provide new insights into the evolution of cell-mediated 258 cytotoxicity. Understanding the molecular pathways initiated by teleost granzymes and death ligands that lead to target cell death, is critical in discovering novel cell death and proliferation pathways. 259
© Copyright 2025 Paperzz