Inflammatory Cytokines and NFκB in Alzheimer’s disease Linda Fisher Stockholm 2006 Doctoral dissertation, 2006 Department of Neurochemistry Arrhenius Laboratories for Natural Sciences Stockholm University, SE-106 91 Stockholm Sweden Linda Fisher, 2006 ISBN 91-7155-265-0 Printed in Sweden by Universitetsservice US-AB Stockholm, 2006 ABSTRACT Alzheimer’s disease is the most common form of dementia in the world. It is a progressive neurodegenerative disorder characterized neuropathologically by extracellular deposition of senile plaques, as well as intracellular neurofibrillary tangles. The main constituent of the senile plaques is the β-amyloid peptide, which is neurotoxic and has been suggested to directly contribute to the progressive neurodegeneration observed in Alzheimer’s disease. Surrounding the senile plaques there are activated astrocytes and microglia. It is believed that these activated glial cells contribute to neurotoxicity through secretion of proinflammatory cytokines, like interleukin-1β and interleukin-6, and induction of oxidative damage. The activation of glial cells and induction of cytokine expression is believed to be an early and important event in the development of Alzheimer’s disease. For many inflammatory actions, including the cytokine induction in glial cells, the transcription factor NFκΒ plays a key role. This suggests that therapeutical strategies aimed to control the development of Alzheimer’s disease could include administration of drugs that hinder NFκΒ activation. The major aim of this thesis was to examine the effects of β-amyloid together with interleukin-1β on cytokine expression as well as NFκB activation in glial cells. In addition, the possibility to block NFκB activation, and downstream effects like interleukin-6 expression, by using a double-stranded oligonucleotide containing the consensus sequence for NFκB was investigated. The possibility to improve the cellular uptake of the oligonucleotide by using a cell-penetrating peptide linked to the oligonucleotide by peptide nucleic acid was also investigated. Further, the uptake and antisense effects of peptide nucleic acid directed against the β-amyloid precursor protein was also investigated. The results obtained provide supportive evidence that inflammatory cytokines are induced by β-amyloid, and that they can indeed potentiate its effects. The results further demonstrate that by blocking the transcription factor NFκB, the induction of interleukin-6 expression can be inhibited. By using an improved cellular delivery system, the uptake of the NFκB oligonucleotide decoy and hence the downstream cytokine inhibition could be increased. The results also show the possibility to downregulate the β-amyloid precursor protein by using peptide nucleic acid. Taken together, these results demonstrate the possibility to decrease the inflammatory reactions taken place in Alzheimer’s disease brains, which may ultimately lead to a possible way of controlling this disorder. LIST OF PUBLICATIONS This thesis is based on the following publications, referred to in the text by the corresponding Roman numerals: Paper I. L. Fisher, U. Soomets, V. Cortés Toro, L. Chilton, Y. Jiang, Ü. Langel and K. Iverfeldt. Cellular delivery of a double-stranded oligonucleotide NFκB decoy by hybridization to complementary PNA linked to a cell-penetrating peptide. Gene Therapy 2004; 11, 1264-1272 Paper II. L. Holmlund*, V. Cortés Toro and K. Iverfeldt. Additive effects of amyloid β fragment and interleukin-1β on IL-6 secretion in rat primary glial cultures. International Journal of Molecular Medicine 2002; 10, 245-250 Paper III. L. Fisher, M. Samuelsson, Y. Jiang, V. Olsson, R. Figueroa, E. Hallberg, Ü. Langel and K. Iverfeldt. Targeting cytokine expression in glial cells by cellular delivery of an NFκB decoy. Submitted Paper IV. L. Adlerz, U. Soomets, L. Holmlund*, S. Viirland, Ü. Langel and K. Iverfeldt. Down-regulation of amyloid precursor protein by peptide nucleic acid oligomer in cultured rat primary neurons and astrocytes. Neuroscience Letters 2003; 336, 55-59 * Maiden name of L. Fisher ADDITIONAL PUBLICATIONS BY L. FISHER M. Samuelsson, L. Fisher and K. Iverfeldt. β-Amyloid and interleukin-1β induce persistent NFκB activation in rat glial cells. International Journal of Molecular Medicin 2005; 16, 449-453 G. Wolf, R. Yirmiya, I. Goshen, K. Iverfeldt, L. Holmlund*, K. Takeda and Y. Shavit. Impairment of interleukin-1 (IL-1) signaling reduces basal pain sensitivity in mice: genetic, pharmacological and developmental aspects. Pain 2003; 104, 471-480 L. Holmlund*, G. Imreh, E. Hallberg and K. Iverfeldt. Real time monitoring of apoptosis, induced by beta-amyloid peptide, in SH-SY5Y neuroblastoma cells expressing a GFP-tagged nuclear pore protein. Miami Nature Biotechnology Short Reports. The Scientific World 2001; 1, 54SR ISSN 1532-2246. Proceeding TABLE OF CONTENTS 1. INTRODUCTION 1.1. Alzheimer’s disease 1.1.1. Neurofibrillary tangles and amyloid plaques 1.1.2. β-Amyloid Precursor Protein processing 1.1.3. Alzheimer’s disease-linked mutations 1.1.4. The amyloid cascade hypothesis 1.1.5. Aggregation of the β-amyloid peptide 1.1.6. β-Amyloid toxicity 1 1 1 2 4 6 7 8 1.2. Cells of the CNS 1.2.1. Astrocytes 1.2.2. Microglia 10 11 12 1.3. Inflammation in Alzheimer’s disease 1.3.1. Role of glial cells in Alzheimer’s disease 1.3.2. Proinflammatory cytokines 1.3.3. The interleukin-1 system 1.3.4. Interleukin-6 1.3.5. Role of inflammatory cytokines in Alzheimer’s disease 12 13 15 16 17 18 1.4. Nuclear Factor κB 1.4.1. The Rel family of proteins 1.4.2. NFκB in neurons and glial cells 1.4.3. Role of NFκB in Alzheimer’s disease 19 21 22 23 1.5. Interference with gene expression 1.5.1. PNA as antisense instrument 1.5.2. Transcription factor decoy 25 26 27 1.6. Cellular uptake of PNA and oligonucleotides 1.6.1. Cell-penetrating peptides 27 27 2. AIMS OF THE STUDY 29 3. METHODOLOGICAL CONSIDERATIONS 30 3.1. Cell cultures 3.1.1. Rinm5F cell line 3.1.2. Primary mixed glial cell cultures 3.1.3. Primary neuronal cell cultures 30 30 30 30 3.2. Design of PNA and transport peptide-PNA constructs 31 3.3. Cell treatments 32 3.4. Analysis of specific protein-DNA interactions 3.4.1 Electrophoretic mobility shift assay 33 33 3.5. Analysis of mRNA expression 3.5.1. RT-PCR analysis 35 35 3.6. Analysis of protein expression 3.6.1. Western blot 3.6.2. ELISA 37 37 38 3.7. Fluorescence microscopy 40 4. RESULTS AND DISCUSSION 42 4.1. 4.2. 4.3. 4.4. 4.5. 4.6. Cellular delivery of NFκ κB decoy IL-1β β - and Aβ β -induced NFκ κB activation IL-1β β - and Aβ β -induced upregulation of cytokine expression Inhibition of NFκ κB-binding activity in glial cells Decreased IL-6 mRNA expression using NFκ κB decoy Downregulation of APP in neurons and glial cells using PNA 42 44 45 47 48 51 5. CONCLUSIONS 53 6. ACKNOWLEDGEMENTS 55 7. REFERENCES 56 8. ORIGINAL PUBLICATIONS 88 ABBREVIATIONS Aβ AD AGE AICD AP AP-1 Aph-1 ApoE APP BACE BBB C/EBP cDNA CGCs CNS CPP ECL ELISA EMSA FPRL1 GFAP HIV-1 HRP HVJ IFN IκB IKK β-amyloid Alzheimer’s disease advanced glycation end products APP intracellular domain alkaline phosphatase activator protein-1 anterior pharynx defective-1 apolipoprotein E β-amyloid precursor protein β-site APP cleaving enzyme blood brain barrier CCAAT enhancer binding protein complementary DNA cerecellar granule cells central nervous system cell-penetrating peptide enhanced chemiluminescence enzyme-linked immunosorbent assay electrophoretic mobility shift assay formyl peptide receptorlike 1 glial fibrillary acidic protein human immunodeficiency virus 1 horseradish peroxidase haemagglutinating virus of Japan interferon inhibitory κB IκB kinase IL IL-1R IL-1ra IL-1RAcP IL-6R iNOS IRAK M-CSF mRNA NFκB NFT NGF NO NSAIDs ODN Pen-2 PNA PS RAGE RHD ROS RPA rRNA RT-PCR siRNA TACE TNFα TP TRITC interleukin IL-1 receptor IL-1 receptor antagonist IL-1 receptor accessory protein IL-6 receptor inducible nitric oxide synthase IL-1 receptor-associated kinase macrophage-colony stimulating factor messenger RNA nuclear factor κB neurofibrillary tangles nerve growth factor nitric oxide nonsteroidal antiinflammatory drugs oligodeoxyribonucleotide presenilin enhancer-2 peptide nucleic acid presenilin receptor for advanced glycation end products rel homology domain reavtive oxygen species ribonuclease protection assay ribosomal RNA reverse transcriptionpolymerase chain reaction short interfering RNA tumor necrosis factor-α converting enzyme tumor necrosis factor α transportan tetramethyl-rhodamine isothiocyanate L. Fisher 1. INTRODUCTION 1.1. Alzheimer’s disease A century ago, in November 1906 to be exact, the neuropathologist and psychiatrist Alois Alzheimer described the first case of dementia, which was later published in 1907 (Alzheimer, 1907). Alzheimer described a 51-year old woman that had developed memory deficits and progressive loss of cognitive abilities. A brain autopsy revealed the classical signs of what is today known as Alzheimer’s disease (AD), i.e. senile plaques and neurofibrillary tangles throughout the neocortex and hippocampus. Today, AD is the most common form of dementia. It is affecting about 10% of the population over the age of 65 and the frequency increases to nearly 50% by the age of 80 (Hof et al., 1995; Smith, 1998). AD is a devastating disease that robs its victims of their most cherished human qualities, the ability to remember, think, reason, and speak. It also leads to death; in fact it is the fourth leading cause of death amongst the elderly. This, together with the fact that our world’s population is ‘aging’, leading to that the number of demented people will increase, emphasizes the need for an effective treatment of AD. In the last decades novel genetic factors and cellular mechanisms, causative of the disease, have been revealed, helping in the search for new therapeutic targets and drugs. 1.1.1. Neurofibrillary tangles and amyloid plaques AD is characterized by cortical atrophy in the form of gyral shrinkage, widening of the sulci, and enlargement of the ventricles. The ‘memory centre’ hippocampus and the entorhinal cortex are the first regions to be affected. As the disease progresses, more regions are affected including the temporal and parietal lobes (Braak and Braak, 1994). Pronounced neurodegeneration, synaptic loss, and gliosis, are also observed in AD brains. The most prominent microscopical alterations are the presence of extracellular senile or amyloid plaques and the intracellular neurofibrillary tangles (NFTs) (Katzman and Saitoh, 1991). NFTs are abnormal, filamentous inclusions found in the cell bodies of neurons, primarily composed of abnormally phosphorylated tau (Grundke-Iqbal et al., 1986; Wischik et al., 1988; Lee et al., 1991; Goedert et al., 1993). Tau, a microtubuleassociated protein, forms paired helical filaments upon hyperphosphorylation, which leads to impaired axonal transport and eventually cell death. The senile plaques are extracellular deposits with a dense central core, surrounded by dystrophic neurites, indicating that a neurodegenerative process is taking place. Activated microglia and astrocytes are also present, indicating an inflammatory reaction. The main constituent of the plaques is the 40 or 42 amino acid long β-amyloid (Aβ) peptide. Two types of plaques have been morphologically distinguished from one another; the neuritic plaques and the diffuse plaques (Glenner and Wong, 1984b). The longer form of Aβ (Aβ42) has been found to be the main residue in the core of the neuritic plaque surrounded by aggregates consisting of both Aβ40 and Aβ42. The neuritic plaques can be stained by classical silver staining, or by the histological amyloid dyes Congo red and thioflavin. By using antibodies directed against Aβ, the early stage, light, amorphous plaque formations, i.e. diffuse plaques, lacking amyloid fibrillization, can also be detected. INTRODUCTION 1 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 1.1.2. β-Amyloid precursor protein processing The Aβ peptides are derived from the β-amyloid precursor protein (APP). APP is an evolutionary highly conserved glycoprotein, ubiquitously expressed throughout the body. Although many functions for APP have been suggested, its physiological role still remains uncertain. APP is an integral membrane protein with a single membranespanning domain, a large extracellular glycosylated N-terminus and a shorter cytoplasmic C-terminal tail. There are three major isoforms of APP with 695, 751, and 770 residues, respectively, that arise due to alternative splicing (Kitaguchi et al., 1988; Ponte et al., 1988; Tanzi et al., 1988). The most abundant form in neurons is APP695, while in astrocytes and microglia the APP751 and APP770 isoforms are preferentially expressed (Haass et al., 1991). Neurons express the highest levels of APP and have also been shown to secrete considerable amounts of Aβ (Haass et al., 1992). However, other brain cells, including astrocytes and microglia, also release the Aβ peptide. APP molecules undergo several specific endoproteolytic cleavages by secretases, resulting in the formation and release of fragments into the vesicle lumens, the cytosol, and the extracellular space. At least two different proteolytic processing pathways exist, which will determine the quantity of the Aβ formed in the cell (Palmert et al., 1989; Haass et al., 1992; Seubert et al., 1993). The mechanism by which the proteolytic cleavage of APP generates Aβ is fairly recognized (Fig. 1). The initial step in this pathway is executed by β-secretase, which cleaves APP at the N-terminal junction of the Aβ sequence. This cleavage results in secretion of an N-terminal fragment of APP, sAPPβ, and formation of a 99-residue C-terminal fragment, C99, left in the membrane (Seubert et al., 1993). BACE (β-site APP cleaving enzyme), a member of the pepsin family of aspartyl proteases, has been identified as the enzyme responsible for the βsecretase cleavage (Hussain et al., 1999; Sinha et al., 1999; Vassar et al., 1999; Yan et al., 1999a). There are two homologues of BACE that have been identified, BACE1 and BACE2. BACE1 is mainly expressed in the brain and most likely responsible for the processing of APP. The final catalytic step in the Aβ production includes γ-secretase cleavage. The Cterminal fragment left in the membrane generated from β-secretase cleavage, C99, can further be processed to produce Aβ. γ-Secretase cleaves the APP fragments within their transmembrane domain. γ-Secretase shows low sequence specificity and generates Aβ peptides differing in length. Cleavage between residues 711 and 712 in the APP 770 isoform generates a 40-residue (Aβ40) peptide and cleavage after 713, a 42-residue (Aβ42) peptide. Aβ40 is the most abundant product, whereas the longer Aβ42 variant is generated to a lesser extent (Wang et al., 1996b). Intense research efforts have, over the past few years, provided new information about γ-secretase. It is recognized as a large multicomponent complex including presenilin (PS), nicastrin, anterior pharynx defective1 (Aph-1), and presenilin enhancer-2 (Pen-2) (Zhang et al., 2000; Chung and Struhl, 2001; Francis et al., 2002). All these components have been reported to be essential for the γ-secretase activity, although PS is thought to contain the catalytic activity, being responsible for the actual cleavage (De Strooper et al., 1998; De Strooper, 2003; Edbauer et al., 2003; Li et al., 2003; Luo et al., 2003). In addition, PS-dependent cleavages at alternative sites that may preceed γ-cleavage have recently been discovered. These include the ε-cleavage, which occurs after position 49 in Aβ (Gu et al., 2001; 2 INTRODUCTION L. Fisher Sastre et al., 2001; Yu et al., 2001) and ξ-cleavage, which cleaves APP after position 46 in the Aβ sequence (Zhao et al., 2004). Although strong evidence points to a crucial role for PS in the generation of Aβ, it should be noted that also PS-independent production of Aβ has been reported (Armogida et al., 2001; Wilson et al., 2002a). Even though two PS homologues have been identified, PS1 and PS2 (Levy-Lahad et al., 1995; Rogaev et al., 1995; Sherrington et al., 1995), most data are based on studies on PS1. Figure 1. Processing of APP. The β-pathway is shown at the top, where APP is cleaved by β- and γ-secretase to generate Aβ. At the bottom the non-amyloidogenic pathway is shown, where cleavage of APP by α- and γ-secretase will result in the formation of the p3 fragment. INTRODUCTION 3 Inflammatory Cytokines and NFκB in Alzheimer’s Disease The most prominent APP processing pathway does not result in Aβ production. In the ‘normal’ pathway, APP is first cleaved by α-secretase, 12 amino acids N-terminal to the transmembrane domain, resulting in the secretion of a soluble N-terminal fragment, sAPPα, into the extracellular space. An 83-residue C-terminal fragment, C83, remains in the membrane. The cleavage by α-secretase occurs between amino acids 16 and 17 within the Aβ sequence, preventing the formation of Aβ. Thus, this pathway is also referred to as the non-amyloidogenic pathway. Suggested candidates for the α-secretase activity are three members belonging to the metalloprotease ADAM family, ADAM-9, ADAM-10, and ADAM-17 or tumor necrosis factor-α converting enzyme (TACE) (Buxbaum et al., 1998; Koike et al., 1999; Lammich et al., 1999; Asai et al., 2003). The membrane-bond fragment C83, produced after α-secretase cleavage, can, like C99, also undergo γ-secretase-mediated intramembrane proteolysis. C83 is cleaved by γ-secretase to produce the non-amyloidogenic p3 peptide. In addition to p3 and Aβ, the recently described C-terminal cytoplasmic fragment of APP, CTFγ, also referred to as AICD (APP intracellular domain), is produced by PS-dependant cleavage of C83 and C99 (Pinnix et al., 2001). 1.1.3. Alzheimer’s disease-linked mutations Most AD cases are sporadic with unknown cause, with suggested risk factors including head trauma, female gender, vascular disease, and low education in addition to old age (Mortimer et al., 1991; Launer et al., 1999). However, about 5-10% of all AD cases account for the familial forms, caused by mutations in genes associated with AD. The two forms usually differ in regard to age of onset, where the familiar form can start as early as in the late 20’s compared to mid 60’s for the sporadic form. Already in the 1960’s, genetic factors were indicated to be involved in AD. It was discovered that patients suffering from Down syndrome, which is caused by triplication of chromosome 21, displayed AD-like symptoms and pathology at an early age (Olson and Shaw, 1969). Later, in 1984, it was further revealed that these patients had high levels of a peptide, identified as the Aβ peptide, within their brains (Glenner and Wong, 1984a). When, in 1987, APP was cloned and found to be located on chromosome 21, researchers recognized that the APP gene was a good candidate responsible for the development of AD (Goldgaber et al., 1987; Kang et al., 1987; Robakis et al., 1987; Tanzi et al., 1987). However, it took another four years until the first mutation in the APP gene was identified as being a specific genetic cause of AD (Goate et al., 1991). Soon after this, several APP mutations were discovered (Tabel 1) (Chartier-Harlin et al., 1991; Fernandez-Madrid et al., 1991; Murrell et al., 1991; Carter et al., 1992; Hendriks et al., 1992; Jones et al., 1992; Kamino et al., 1992; Mullan et al., 1992; Peacock et al., 1993). Most of these mutations are located close to one of the three secretase cleavage sites on APP, promoting the generation of Aβ by favoring proteolytic processing of APP by β- or γ-secretase (Citron et al., 1992; Cai et al., 1993; Suzuki et al., 1994). Moreover, APP mutations found within the Aβ sequence enhance the aggregation of Aβ into fibrils (Wisniewski et al., 1991; Nilsberth et al., 2001). Nevertheless, AD-linked mutations could only explain the genetic cause of AD in a small fraction of families suffering from familial AD. There were still families with apparent familial AD where APP gene mutations had not been found. In 1995, the first mutation in a gene on chromosome 14 4 INTRODUCTION L. Fisher was identified as a causative AD mutation (Sherrington et al., 1995). Today, more than 150 mutations have been identified in this gene coding for PS1. In addition, ten mutations in the PS2 gene on chromosome 1 have been identified as AD-linked mutations (see AD mutations database). All AD causative mutations discovered so far are located in these three genes, and most of them have been reported to increase the generation of Aβ through effects on APP processing (Citron et al., 1992; Suzuki et al., 1994; Scheuner et al., 1996; St George-Hyslop, 2000). Even though most cases of AD are sporadic, the familial AD mutations have provided powerful insights into the underlying mechanism of AD. The clinical picture and the morphological end stage in the brain of individuals with sporadic or familial AD certainly appear to be the same even if the etiology differs. Table 1. AD associated APP mutations. Name Swedish Flemish Artic Dutch Italian Iowa Austrian Iranian French German Florida London Indiana Australian Mutation Lys⇒Met/Asn⇒Leu Asp⇒Asn Ala⇒Gly Glu⇒Gly Glu⇒Gln Glu⇒Lys Asp⇒Asn Thr⇒Ile Thr⇒Ala Val⇒Met Val⇒Ala Ile⇒Val Ile⇒Thr Val⇒Ile Val⇒Phe Val⇒Gly Val⇒Leu Leu⇒Pro Codon 670/671 678 692 693 693 693 694 714 714 715 715 716 716 717 717 717 717 723 In addition to these AD-linked mutations, other genetic risk factors have been associated with AD. The Apolipoprotein E (ApoE) isoform E4 has been shown to increase the probability for developing AD (Corder et al., 1993). Studies on knockout and transgenic mice suggests that this particular isoform of ApoE promotes Aβ deposition and aggregation (Bales et al., 1997; Holtzman et al., 2000). Polymorphism in several inflammatory genes has also been linked with increased risk for developing AD. The possible involvement of the inflammatory cytokines interleukin (IL)-1 and IL-6 in AD will further be discussed in section 1.3.5. INTRODUCTION 5 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 1.1.4. The amyloid cascade hypothesis The amyloid cascade hypothesis is currently the most favored model explaining the pathogenic events causing AD (see Fig. 2). It is supported not only by the fact that most AD mutations increase the production of Aβ, especially Aβ42, but also by several studies showing that the levels of Aβ deposits correlates with cognitive decline and the severity of the disease (Cummings et al., 1996; Hsiao et al., 1996; Naslund et al., 2000; Gordon et al., 2001). The hypothesis, first presented in 1991, is based on the postulation that an increased amyloid accumulation provides the driving force for the pathogenesis of the disease leading to the aggregation and formation of senile plaques followed by Altered APP processing inflammatory responses, synapse loss, ⇓ NFTs, neuronal cell death and finally Increased Aβ production and dementia (Selkoe, 1991; Hardy and accumulation Higgins, 1992). Further, the theory states ⇓ that the increased amyloid deposition is Aggregation of extracellular caused due to mismetabolism of APP or Aβ into plaques failure of Aβ clearance. ⇓ Other AD hypotheses put tau and Inflammatory response tangles in focus, stating that in AD the with glial cell activation and normal role of tau in stabilizing cytokine release microtubules is impaired, due to changes ⇓ in the conformation and phosphorylation Progressive synaptic and neuritic injury, of tau (Gray et al., 1987; Lovestone and disruption of neuronal ionic Reynolds, 1997). This hypothesis homeostasis, however, has not gained as much oxidative injury support as the amyloid cascade ⇓ hypothesis. One reason is that not only Altered kinase/phosphatase activities AD, but also other dementias like ⇓ Parkinson’s disease and frontotemporal NFT formation, lobe dementia, demonstrate tau neuronal dysfunction and death pathology (Heutink, 2000; Lee, 2001). ⇓ The absence of plaque pathology in Dementia patients with mutations in tau, causing frontotemporal lobe dementia, further Figure 2. The array of pathogenic events argues against the idea that NTFs would leading to AD as suggested by the amyloid cause plaque formation. Moreover, cascade hypothesis. tangle formation seems to follow Aβ deposition since tau deposition is increasing in transgenic mice overexpressing both human mutant tau and mutant APP, as compared to mice expressing tau mutant alone (Lewis et al., 2001), where no alteration in plaque formation is detected (Lewis et al., 2000). In addition, injection of fibrillar Aβ42 has been shown to enhance the tangle formation in the tau transgenic mice (Gotz et al., 2001). Studies have further demonstrated that Aβ influences tau phosphorylating enzymes in vitro (Rank et al., 2002) and in vivo (Tomidokoro et al., 2001). Recently, a triple transgenic mouse model overexpressing mutant APP and mutant tau on a PS1 mutant knock-in background further showed that senile plaque depositions preceded tau pathology (Oddo et al., 2003). In yet 6 INTRODUCTION L. Fisher another study, it was found that whenever Aβ aggregates were detected in the entorhinal cortex, tau pathology was also found. The opposite was not true as cases were found with advanced tau pathology and no trace of Aβ aggregates (Delacourte et al., 2002). Furthermore, a study from 1996 demonstrated that aged non-demented individuals, with minor cognitive impairments, demonstrated neocortical Aβ deposits, but no tangle formation deficits (Morris et al., 1996). These patients were probably in an early stage of AD, in which the underlying disease process had begun, but had not yet generated sufficient clinical. Together these data suggest that Aβ accumulation is more associated with the primary cause of the disease, while the NFTs may rather be a consequence. This is further supported by a recent report where AD brains with different degrees of severity were studied by immunohistochemical methods, using antibodies against Aβ and paired helical filaments of tau. They found that intracellular Aβ deposition was detected prior to the appearance of paired helical filaments (Fernandez-Vizarra et al., 2004). Even though the amyloid cascade hypothesis is convincing, there remain areas of doubt. Observatios have been made both in mice and humans, that are difficult to reconcile with the hypothesis. For example, there are concerns that the role of NFTs and inflammatory responses is not fully explained by this theory (McGeer and McGeer, 1998; Lee, 2001). Schwab and colleagues recently pointed out that transgenic mice overexpressing Aβ are incomplete models of AD, where little NFTs and activated microglia are seen as opposed to AD (Schwab et al., 2004). In addition, transgenic mice displaying progressive Aβ deposition do not show clear-cut neuronal loss (Games et al., 1995; Hsiao et al., 1996; Irizarry et al., 1997). This could be explained by high levels of the neuroprotective sAPPα, which may protect the brain against Aβ-induced neuronal death in transgenic mice overexpressing mutant forms of APP (Stein et al., 2004). Other reasonable explanations may also include the fact that there are little or no inflammatory mediators like certain cytokines, or human tau molecules in these mice models. Moreover, it has been demonstrated that in contrast to AD amyloid plaque deposits, the accumulated Aβ peptides in the transgenic mice are soluble in certain solutions (Kalback et al., 2002). Thus, differences in disease evolution and biochemistry must be considered when using transgenic animal models to appraise the cause and consequence in AD. 1.1.5. Aggregation of the β -amyloid peptide The 4 kDa Aβ peptide was first identified in 1984 when it was purified from the brain of a patient suffering from Down syndrome (Glenner and Wong, 1984a), as previously mentioned. A year later, it was recognized as the primary component of the senile plaques from AD patient brain tissue (Masters et al., 1985). It has since then been at the center of attention as the major possible cause for AD. The term amyloid means “starch” or “cellulose-like” and its fibrils form so-called cross-β-pleated sheet structures (Eanes and Glenner, 1968; Sunde and Blake, 1998). Amyloid fibrils are long twisted filaments, 6-8 nm wide, resistant to proteolytic degradation. The main Aβ peptides consist of 40 or 42 amino acid residues, where the N-terminal 28 residues, before cleavage from APP, are extracellular and the remaining residues are located within the transmembrane domain (see Fig. 3). Aβ40 accounts for about 90% of all Aβ normally released from cells (AsamiOdaka et al., 1995) along with Aβ42 being the most predominant form found in the senile plaques (Lippa et al., 1998). The longer Aβ42 is more prone to form aggregates as INTRODUCTION 7 Inflammatory Cytokines and NFκB in Alzheimer’s Disease compared to the shorter Aβ peptides. It forms more stable aggregates (Burdick et al., 1992) more rapidly (Jarrett et al., 1993). It is furthermore believed that Aβ42 fibrils promote the aggregation of Aβ40 (Harper and Lansbury, 1997). Thus, a shift to a higher proportion of the Aβ42 may be crucial to the earliest stages of fibril deposition into plaques. Various oligomeric assembly states of Aβ, preceding Aβ fibrils, have been identified, including small soluble oligomers and larger unsoluble protofibrils (Roher et al., 1996; Harper et al., 1997; Walsh et al., 1997; Lambert et al., 1998). The assembly of the Aβ peptide depends on both pH and the concentration of the Aβ peptide. Allthough several Aβ peptides have been demonstrated to form Aβ fibrils, only the fragments including the hydrophobic amino acids 29-35 of the Aβ sequence are known to form stable aggregates at a neutral pH in vitro (Burdick et al., 1992). It has further been reported that peptides lacking the 25-27 amino acid region form aggregates to a lesser extent than Aβ fragments including this sequence. It was suggested that the expected βturn region of Aβ is located within the 25-29 sequence, which in turn contributes to the folding and stability of the Aβ aggregates (Hilbich et al., 1991). Taken together, these data suggest that fibril formation of the Aβ peptide is dependent on the Aβ25-35 region, and that it is this region, in the full length Aβ peptide, that arranges the β-sheet structure. Figure 3. The Aβ sequence, the Aβ25-35 region is shown in dark. 1.1.6. β-Amyloid toxicity Aβ peptides have been shown to be neurotoxic in vitro (Pike et al., 1991b; Pike et al., 1993; Lambert et al., 1998) and in vivo (Kowall et al., 1992; Geula et al., 1998; McKee et al., 1998; Weldon et al., 1998), and it is widely believed that Aβ deposits directly contribute to the progressive neurodegeneration observed in AD (Pike et al., 1992; Rush et al., 1992). It has been suggested that the Aβ peptides exert neurotoxic effects either directly (Scorziello et al., 1996), or by enhancing neuronal vulnerability to excitatory amino acids (Mattson et al., 1992). The toxic properties of Aβ, however, depend on several factors and still hold a lot of debate. It was first reported that it was the aggregated form of Aβ that was directly toxic to cultured neurons, whereas the soluble form did not show any neurotoxic properties at all (Frautschy et al., 1991; Pike et al., 1991a; Pike et al., 1993; Lorenzo and Yankner, 1994; Iversen et al., 1995). Later, it was shown that protofibrils of Aβ as well were neurotoxic (Lambert et al., 1998; Hartley et al., 1999; Walsh et al., 1999). These protofibrils are neither small oligomers nor fullfledged aggregates, but rather intermediates of Aβ on their way from monomeric Aβ 8 INTRODUCTION L. Fisher forming aggregates. Recently, also soluble oligomers of Aβ were shown to be toxic and even suggested to be the most toxic form of Aβ (Klein et al., 2001; Dahlgren et al., 2002; Walsh et al., 2002). Soluble dodecameric assembleys of Aβ have very recently been suggested to be the complex responsible for the memory loss observed in AD at early stages (Lesné et al., 2006). This leaves us still wondering which form (fibrils, protofibrils, oligomers or monomers) of Aβ species is most toxic and has most deleterious effects. It is possible that the large fibrillary Aβ aggregates represent inactive reservoirs of species that are in equilibrium with the smaller, non-fibrillar, putatively neurotoxic assemblies of Aβ. The fact that transgenic mice expressing both mutant APP and PS1 show increased Aβ42 production and cognivive deficits prior to fibrillar amyloid deposition (Holcomb et al., 1998) supports this idea. The findings that levels of soluble Aβ rather than insoluble Aβ fibrils, correlate better with AD severity (Lue et al., 1999) further suggests that non-fibrillar Aβ may be the toxic agent. Some researchers even go as far as to state that the extracellular Aβ fibril deposits are even protective (Davis and Chisholm, 1997; Smith et al., 2002; Lee et al., 2004). It is also worth mentioning that the Aβ peptide has not consistently been demonstrated to be neurotoxic (Vandenabeele and Fiers, 1991). Aβ toxicity may also be a specific pathological response of the aging brain, since intracerebral injection of fibrillar Aβ resulted in profound neuronal loss, tau phosphorylation, and microglial proliferation in the aged, but not in the young adult rhesus monkey brain (Geula et al., 1998). In regards to what Aβ species is most toxic, studies point at Aβ42 to be the most potent out of the Aβ fragments (Zhang et al., 2002). However, the question remains to be answered, if neurotoxic in AD, what type and form of Aβ exert the toxicity? Another question still remains to be answered; how does Aβ exert its toxic effects and cause neuronal degeneration? One suggestion is that it may act through a cell membrane receptor to stimulate intracellular processes, including the hyperphosphorylation of tau, leading to neurodegeneration (Yankner, 1996). In a similar way, it has been suggested that Aβ binds to the receptor for advanced glycation end products (RAGE), causing augmented intracellular oxidative stress and the release of inflammatory factors (Yan et al., 1996; Yan et al., 1999b; Schmidt et al., 2000). RAGE was first discovered due to its ability to bind advanced glycation endproducts (AGEs), which are formed during oxidative stress (Neeper et al., 1992), and is found at elevated levels in neurons close to Aβ deposits and senile plaques (Yan et al., 1996; Sasaki et al., 2001). However, it has been suggested that RAGE is not required for Aβ toxicity, since neuronal cell lines and rat cortical neurons not expressing RAGE, are also vulnerable to Aβ toxicity (Liu et al., 1997). Other receptors that have been identified to interact with Aβ include; the serpin enzyme complex receptor (Boland et al., 1995; Boland et al., 1996), the α-7 nicotinic acetylcholine receptor (Wang et al., 2000; Liu et al., 2001), and the p75 neurotrophin receptor (Yaar et al., 1997). On the contrary, several studies suggest that Aβ itself, not through any receptor, is capable of producing free radicals and reactive oxygen species (ROS) (Hensley et al., 1994; Harris et al., 1995a) or can cause a harmful elevation of intracellular calcium levels (Koh et al., 1990; Mattson et al., 1992; Mogensen et al., 1998). Furthermore, there is evidence that Aβ alters cellular ionic activity, either through interaction with existing ion channels, or by de novo channel INTRODUCTION 9 Inflammatory Cytokines and NFκB in Alzheimer’s Disease formation (Fraser et al., 1997). Metal ions have also been shown to potentiate the neurotoxicity of human Aβ in neuronal cultures by generating ROS (Huang et al., 1999; Cuajungco et al., 2000; Rottkamp et al., 2001). Intracellular Aβ has been observed in differentiated neuronal cell lines (Wertkin et al., 1993; Turner et al., 1996), APP transgenic mice (Wirths et al., 2001; Blanchard et al., 2003; Oddo et al., 2003; Shie et al., 2003), and in affected brain regions of AD patients (Gouras et al., 2000; Fernandez-Vizarra et al., 2004). Due to these observations, together with the fact that deficits may occur before plaque deposition, it cannot be ruled out that the early pathological changes observed in AD are due to intraneuronal Aβinduced toxicity. This is supported by a study showing that neuronal loss in an APP transgenic mouse model did not correlate with the amount of extracellular Aβ deposits, but rather with the high levels of intraneuronal Aβ (Schmitz et al., 2004). This may also explain the poorly understood relationship between senile plaques and NFTs. It is possible that the plaques are not causing tau pathology, but rather the soluble intraneuronal Aβ. In a recent paper, Marchesi discusses even a third possibility for Aβ peptides to exert toxicity (Marchesi, 2005). He speculates that Aβ peptides could, after secretion into the extracellular space, reinsert themselves into the lipid bilayer or that they may, after cleavage of APP, remain in the membrane. These intramembranous Aβ peptides may then exert their potentially toxic effects by creating channel-like structures in the membrane. The Aβ causing neurodegeneration may moreover occur via activation of the inflammatory cells of the brain, i.e. astrocytes and microglia. This will further be discussed in section 1.3. That the Aβ peptide is toxic is well documented, however, it may not only be a dangerous and unfortunate byproduct of APP processing. Aβ is also produced, in small quantities, in healthy individuals under normal physiological conditions. Still today, no normal physiological role has been acknowledged for the Aβ peptide in the brain. Recently, it was suggested that Aβ, in fact, could mediate a physiological homeostatic mechanism in which it reduces excitatory transmission in response to neuronal activity (Kamenetz et al., 2003). Thus, Aβ may act as an endogenous regulator keeping the levels of neuronal activity in check. Aβ has also been suggested to play a role in cholesterol regulation (Puglielli et al., 2005). 1.2. Cells of the CNS The central nervous system (CNS) is composed of two kinds of specialized cell populations, neurons and glial cells. Neurons are the most important cells of the nervous system. They process all of the information that flows through, to, or out of the CNS. This includes the cognitive information through which we are able to think and reason, the sensory information through which we are able to see, hear and touch, and the motor information through which we are able to move. Neurons provide the system with information by receiving messages from the surrounding environment and from each other through electrical impulses and different chemical messengers i.e. neurotransmitters. 10 INTRODUCTION L. Fisher Glial cells, commonly called neuroglia or simply glia, provide support and protection for neurons. There are three types of glial cells in the CNS; astrocytes, microglia and oligodendrocytes. In early postnatal life, oligodendrocytes associates with the axons of nearby neurons to create a myelin sheath. The principal function of the myelin sheath is to provide insulation to the axon to allow for a more efficient conduction of nerve impulses. Other functions of glial cells, mainly attributed astrocytes, are to serve primarily as physical support for neurons, keeping them in place and to supply nutrients and chemicals needed for proper functioning of the neurons. Microglia cells, on the other hand, have been implicated in the important function in removing and cleaning up the debris in the brain. 1.2.1. Astrocytes Astrocytes are the most abundant type of glial cell within the CNS. They were first visualized over a century ago (Andriezen, 1893), characterized by an oval nuclei and a large star-shaped morphology with many fine processes radiating in all directions. These processes contain a specific form of cytoskeletal intermediate filament called glial fibrillary acidic protein (GFAP) (Bignami et al., 1972). GFAP is exclusively found in astrocytes, thus serving as a suitable marker for identification of these cells. There are two subtypes of astrocytes, the type-1 astrocyte that originates in embryonic life and the type-2 astrocyte that comes from the oligodendrocyte-type-2 astrocyte progenitor that arises early in postnatal life (Raff et al., 1983; Raff, 1989). The protoplasmic type-1 astrocyte has thicker branches and is most evident in the grey matter of the brain, whereas the fibrous type-2 astrocyte has longer processes and is predominantly found in the white matter (Lillien and Raff, 1990). Astrocytes have been attributed many different functions, including cellular support during CNS development. In the developing brain, they form a structural framework to guide the migration of developing neurons to their final position in the brain (Silver and Sapiro, 1981; Hatten, 1985; Hatten, 1990). Furthermore, they produce numerous molecules that have been implicated in the positive support of axon growth during development (Liesi and Silver, 1988; Tomaselli et al., 1988; Serafini et al., 1996; Lee et al., 1999). Astrocytes have also been shown to have an important role in the formation and maintenance of the blood brain barrier (BBB) (Goldstein, 1987; Janzer and Raff, 1987). The capability of astrocytes to take up neuronally released potassium ions and glutamate from the extracellular space assures the maintenance of physiological extracellular ion homeostasis (Barres, 1991). Another role for astrocytes is to repair damaged areas in the brain caused by injury or infection. Dead neurons are replaced by proliferating astrocytes, which fill the gaps, a process termed gliosis. The astrocytes will form a so-called glial scar and this part of the brain will lose the ability to send and receive nerve impulses (Reier and Houle, 1988). Astrocytes responding to an insult of the CNS are referred to as ‘reactive astrocytes’, and they will remarkably change their morphology (see Fig. 4), motility, function and molecular production. This, in turn, may contribute to an inhibitory and nonsupportive environment for neurons. In fact, the reparation by astrocytes, of damaged areas in the brain, is believed to prevent the regeneration of neuronal processes (Fitch et al., 1999). This means that astrocytes found in the damaged part of the brain, the reactive astrocytes, are inhibitory for growth, while INTRODUCTION 11 Inflammatory Cytokines and NFκB in Alzheimer’s Disease astrocytes in undamaged tissue are accommodating the growth. The consequences and function of reactive astrocytes will be further discussed in section 1.3.1. 1.2.2. Microglia Microglia are the smallest of the glial cells. They are of monocytic origin and invade the developing brain during embryonic and early postnatal life (Jordan and Thomas, 1988; Hickey et al., 1992; Ling and Wong, 1993). The microglia cells serve as the brain’s immune cells. They are basically specialized macrophages unique to the CNS, where their function is to destroy invading microorganisms and to clear dead neurons by phagocytosis. However, in the mature brain, microglia are normally found in a ramified (resting) state, lacking phagocytotic activity. At this state they also show very low or undetectable levels of membrane ligands and receptors essential for mediating or inducing typical macrophage functions (Kreutzberg, 1996). To carry out these kinds of functions, microglia need to go through a morphological change of the cell body, processes and the cell surface protein expression. This activation occurs in response to stress stimuli like CNS infection, inflammation or injury. In their activated form, microglia are not only phagocytotic, but also secretory cells with a wide range of secreted molecules, most of which are involved in functions such as tissue remodeling and immune response including IL-1β (Giulian et al., 1988b), nerve growth factor (NGF) (Mallat et al., 1989) and IL-6 (Frei et al., 1989; Dickson et al., 1993). There is very little known about the function of resting microglia. Maybe, as macrophages do in other tissues, they contribute to homeostasis in the CNS, or maybe they are just simply there waiting for something to happen. 1.3. Inflammation in Alzheimer’s disease Compelling evidence gained over the last decade has supported the idea that inflammation is associated with AD development and pathology. Still, it is nothing new that inflammation may take part in this destructive disorder. In fact, already in 1907, Alois Alzheimer noticed signs of an inflammatory reaction in the dementing disease he described (Alzheimer, 1907). Inflammatory conditions such as head trauma and infection have been reported as potential risk factors for AD (Mortimer et al., 1991; Breteler et al., 1992). There is also evidence stating that inflammatory mechanisms occur in the brain regions exhibiting high levels of AD pathology, yet are absent in brain regions that are spared from AD pathology (Akiyama et al., 2000). In addition, numerous epidemiological studies have shown that chronic use of nonsteroidal anti-inflammatory drugs (NSAIDs) significantly decreased the risk of developing AD (Rogers et al., 1993; Breitner et al., 1994; Breitner et al., 1995; Breitner, 1996; McGeer et al., 1996; Stewart et al., 1997; in t' Veld et al., 2001; Pasinetti, 2002; Hoozemans et al., 2003), suggesting a correlation between inflammation and AD. Hence, brain inflammation has become one of the major focuses for AD research. The role of inflammation in AD though is yet to be elucidated, whether it is a secondary process or directly contributes to the disease process is still unclear. However, the brain inflammatory response is known to contribute to the development of tissue injury during other types of neurodegenerative conditions, 12 INTRODUCTION L. Fisher indicating that anti-inflammatory treatment can also reduce the brain damage occurring in AD. A B Figure 4. Morphology of A non-treated primary mixed glial cultures containing both astrocytes (white arrow) and microglia (black arrow)and B primary mixed glial cultures treated with Aβ with astrocytic processes (white arrow) and clusters of microglia (black arrow). 1.3.1. Role of glial cells in Alzheimer’s disease The major players involved in the inflammatory processes in AD are thought to be microglia and astrocytes, although cells of the immune system are also likely to take part. Aβ has been shown to attract and activate microglia leading to clustering of these cells around Aβ deposits (cf., Fig. 4) (McGeer et al., 1988; Haga et al., 1989; Griffin et al., 1995; Dickson, 1997; Sasaki et al., 1997). The occurrence of activated microglia surrounding Aβ deposits may point to a phagocytic attempt by microglia to remove Aβ plaques. Several studies have demonstrated microglial phagocytosis of Aβ (Frautschy et al., 1992; Paresce et al., 1997; Kopec and Carroll, 1998; Mitrasinovic and Murphy, 2003). Failure of microglial Aβ clearence has been proposed to contribute to AD pathology. Morphometric studies of Aβ plaques in humans and in APP transgenic mice have shown that an increased number of microglia does not result in a corresponding increase in Aβ deposit degradation (Wegiel et al., 2001; Wegiel et al., 2003). The fact that it rather correlates with the continued growth of the Aβ deposit and total plaque volume suggests that the microglia are not able to internalize and degrade fibrillar Aβ, but may rather concentrate it. But why would microglia no longer be able to clear the brain from Aβ in this disease? Aged glial cells show signs of an impaired phagocytic activity (Yu et al., 2002). This may lead to an overload of Aβ deposits which in turn may simply be too hard for microglia to digest. An additional factor that can be of importance is the upregulation of potent phagocytosis inhibitors, like glucocorticoids and prostaglandins, observed in AD (Hutchison and Myers, 1987; Russo-Marie, 1992; Chao et al., 1994; Roldan et al., 1997; von Zahn et al., 1997). Activation of microglia and astrocytes by Aβ also leads to an increased secretion of proinflammatory cytokines like IL-1β and IL-6, as well as chemokines, and complement factors (Lee et al., 1993; Del Bo et al., 1995; Eriksson et al., 1998; Johnstone et al., 1999; Akama and Van Eldik, 2000; Toro et al., 2001; paper II). In INTRODUCTION 13 Inflammatory Cytokines and NFκB in Alzheimer’s Disease addition, activated microglia also produce oxygen- and nitrogen- free radicals. Thus, microglia may play an important role in AD pathogenesis via Aβ-induced expression of inducible nitric oxide synthase (iNOS) (Ii et al., 1996; Weldon et al., 1998) and production of ROS (El Khoury et al., 1996; Klegeris and McGeer, 1997). Products released by activated microglia may be the mechanism behind the reported neurotoxicity that microglia displays (Klegeris et al., 1994). The role of microglia in neuronal degeneration in response to Aβ is further supported by studies demonstrating microgliadependent Aβ-induced neurotoxicity (Giulian et al., 1996; Chen et al., 2005). Additionally, in vivo imaging studies have shown that accumulation of activated microglia in AD pathology brain areas occurs at a relatively early stage of the disease process, probably before neurodegenerative changes occur (Cagnin et al., 2001). In the enthorinal and frontal cortex, activated microglia showed a higher correlation with synapse loss than NFTs and Aβ deposits (Lue et al., 1996), suggesting that activated glial cells may be a prime inducer of the neuronal damage that takes place in AD. Moreover, diffuse Aβ deposits, sometimes found in non-demented elderly individuals, lack activated microglia in contrast to the Aβ deposits found in AD patients (Mackenzie et al., 1995), pointing to an important role for microglia in the initiation of plaque progression, neuritic pathology and the initiation of AD development itself. An involvement of microglia in NFT formation has, in addition, been demostrated (Kitazawa et al., 2005). Many different mechanisms have been proposed for Aβ activation of microglia. Microglia can express scavenger receptors through which Aβ can mediate the secretion of ROS via an NFκB-mediated mechanism (El Khoury et al., 1996; Bales et al., 1998; Coraci et al., 2002). Both class A and class B scavenger receptors are found upregulated in association with the senile plaques of AD brains (Christie et al., 1996; Ricciarelli et al., 2004). The G-protein-coupled chemoattractant receptor, formyl peptide receptor-like 1 (FPRL1), has also been suggested to mediate the activation of microglia observed in AD (Le et al., 2001; Yazawa et al., 2001; Cui et al., 2002). Additionally, a cell surface receptor complex, including the scavenger receptor CD36, the integrin-associated protein CD47, and the α6β1-integrin receptor has also been shown to mediate microglial activation by fibrillary Aβ (Bamberger et al., 2003). Moreover, RAGE has also been shown to be a receptor that can bind Aβ and trigger signals leading to cellular activation, inflammatory cytokine production, and ROS generation in microglia (Yan et al., 1999b; Lue et al., 2001; Wyss-Coray et al., 2001). Binding of Aβ to neuronal RAGE have been demonstrated to induce the expression of macrophage-colony stimulating factor (M-CSF) (Du Yan et al., 1997), which then in turn can activate microglia (Stanley et al., 1997). The role of astrocytes in the inflammatory process associated with AD is not really elucidated. It has been suggested that the activation of astrocytes is a secondary consequence of microglia activation (Lee et al., 1993; Frautschy et al., 1998). Reactive astrocytes are also found in the vicinity of the senile plaques (Duffy et al., 1980; Dickson et al., 1988; Pike et al., 1995a), and they have been proposed to support microglia phagocytosis and degradation of Aβ (Yamaguchi et al., 1998; Thal et al., 2000; Nagele et al., 2003; Wyss-Coray et al., 2003). This has been suggested to occur through a RAGEdependent process (Sasaki et al., 2001). The amount of Aβ accumulation within activated astrocytes has also been shown to correlate with the severity of local AD pathology (Nagele et al., 2003). However, the phagocytotic ability by astrocytes is disputed, since 14 INTRODUCTION L. Fisher APP transgenic mouse models have also provided evidence that two of the astrocyte protein products, ApoE and α1-ACT, play an important role in amyloid plaque deposition (Bales et al., 1997; Mucke et al., 2000; Nilsson et al., 2001). One might argue that the astrocytic Aβ could be produced intracellularly. However, since the expression of APP in astrocytes is low, the internal production of Aβ is unlikely to be the major source of the accumulated Aβ found within these cells. The debris-clearing activity of degenerated neurons that might contain Aβ may explain the source of astrocytic Aβ rather than their ability to phagocytose the actual peptide. The main function of reactivated astrocytes in the CNS is thought to be associated with the release of proinflammatory products, which can contribute to neuronal cell damage. Reactive astrocytes, or gliosis, results in an increased expression of proinflammatory cytokines which, in turn, has been shown to stimulate the activation and proliferation of astrocytes (Giulian and Lachman, 1985; Giulian et al., 1988a; Selmaj et al., 1990; Merrill, 1991). This set up for a positive feed-back loop, where more reactivated astrocytes secrete more cytokines that further activate more astrocytes. Reactive astrocytes are co-localized with diffuse plaques in the absence of dystrophic neuritis in the early stages of AD, suggesting that plaque-induced gliosis is an early event in the disease, possibly contributing to AD pathology. Along with microglia, astrocytes as well have been shown to increase the levels of iNOS (Akama and Van Eldik, 2000; Simic et al., 2000), which may lead to nitric oxide (NO) production and further neuronal damage. Since the microglia and the astrocytes can have both neuroprotective functions by degrading extracellular Aβ and secrete neurotrophic factors, and neurodegenerative functions by secreting proinflammatory cytokines and cytotoxic agents, it is difficult to determine their role in the AD process. It is possible that microglia and astrocytes exhibit different roles in plaque evolution. Astrocytes may contribute to amyloid deposits whereas microglia may have a role in the clearance of the plaques. Studies showing that astrocytes inhibited the microglial ability to ingest plaques or Aβ in vitro support this idea (Shaffer et al., 1995; DeWitt et al., 1998). So maybe it is not the failure of microglia to phagocytose Aβ, but rather the inhibiting function of activated astrocytes that results in the increased levels of Aβ deposits in AD. However, a recent report suggests that astrocytes can rescue neurons from apoptosis induced by activated microglia exposed to Aβ (von Bernhardi and Eugenin, 2004). On the other hand, they could not detect this astrocyte modulation when proinflammatory factors where added. In their report, they suggest that activated astrocytes can decrease microglial reaction under physiological conditions, whereas under proinflammatory conditions, this downregulation by astrocytes may fail and thereby enhance microglial activation, secretion of inflammatory cytokines, and cytotoxicity. Anyhow, it seems like both cell types can contribute to the inflammatory reaction and neuronal damage associated with AD by secreting proinflammatory cytokines and oxygen free radicals. 1.3.2. Proinflammatory cytokines Cytokines are a family of proteins known to regulate the intensity and duration of immune and inflammatory responses (Dinarello, 1989; Benveniste, 1992). The constitutive expression of cytokines and their receptors in the CNS is fairly low in healthy tissue, but can be rapidly induced by a variety of endogenous or exogenous stimuli including tissue injury, stress and immune challenge. This to serve an array of INTRODUCTION 15 Inflammatory Cytokines and NFκB in Alzheimer’s Disease immune signaling and effector functions. The synthesis of cytokines is increased in inflammatory states, where they mediate their biological effects by binding to specific cell surface receptors on target cells (Araujo et al., 1989; Holliday and Gruol, 1993; Chao et al., 1995; Skaper et al., 1995). Due to differences in biological activity, cytokines have been categorized as pro- or anti-inflammatory. The anti-inflammatory cytokines include IL-4, IL-10, IL-13, IL-1 receptor antagonist (IL-1ra) and transforming growth factor β, and they act either by inhibiting the expression or reversing the effects of proinflammatory cytokines. The proinflammatory cytokines include IL-1, IL-6 and tumor necrosis factor α (TNFα). However, the classification of the different cytokines is not straightforward. Both IL-6 and TNFα, for instance, have also displayed antiinflammatory properties, and/or neuroprotective properties (Hama et al., 1991; Gadient and Otten, 1997; Akiyama et al., 2000; Tarkowski et al., 2003). In addition to microglia and astrocytes, neurons are also able to produce proinflammatory cytokines (Tchelingerian et al., 1996; Li et al., 2000; Friedman, 2001). This production may further trigger inflammatory processes that worsen the environment and lead to neuronal toxicity and death. 1.3.3. The interleukin-1 system IL-1 is a family of three closely related proteins, IL-1α, IL-1β and IL-1ra. They are all products of separate genes. The two agonists, IL-1α and IL-1β, are synthesized as precursor proteins, and IL-1β needs to undergo proteolytic cleavage to generate the mature and active cytokine (March et al., 1985). After proteolytic maturation, they exert their actions by binding to the IL-1 receptor on the cell surface. There are two IL-1 receptors known today that bind IL-1α and IL-1β, the IL-1 receptor type I and II (IL-1RI and IL-1RII) (Sims et al., 1988; McMahan et al., 1991). IL-1RI is thought to be the one receptor mediating all the IL-1 signal transductions, whereas IL-1RII, lacking the intracellular domain, is believed to work as a negative regulator of IL-1 signaling i.e. a decoy receptor (Colotta et al., 1993; Sims et al., 1993). In addition, IL-1RI requires association with an accessory protein (IL-1RAcP) for signal transduction (Greenfeder et al., 1995). The antagonist, IL-1ra, binds to IL-1RI without initiating the association with the IL-1RAcP to the receptor, thus blocking the actions of IL-1α and IL-1β (Hannum et al., 1990). The physiological role of IL-1ra is not completely known, but it is probable that its main function is to regulate the action of IL-1. Many actions have been ascribed IL-1 on neurons and glial cells. Activation of a number of second messenger systems has been shown to take place upon IL-1 binding to IL-1RI. Many of them include phosphorylation and activation of transcription factors that leads to the induction of genes important for the immune response, such as acute-phase proteins, iNOS, and other inflammatory cytokines (O'Neill and Kaltschmidt, 1997). The promoter region of the IL-1 gene contain consensus sequences for binding of different transcription factors, such as CCAAT enhancer binding protein (C/EBP), nuclear factor κB (NFκB) and activator protein-1 (AP-1) (Furutani et al., 1986; Shirakawa et al., 1993). Acting through these types of transcription factors, IL-1 has been demonstrated to induce not only other cytokines like IL-6 and TNFα, but also its own production (Chung and Benveniste, 1990; Sparacio et al., 1992; Boutin et al., 2001). During the CNS host defense, IL-1β has been proposed to act as a reporter for the immune system, informing the nervous system about the state of function by controlling sickness behavior, fever and 16 INTRODUCTION L. Fisher neuroendocrine changes (Dinarello, 1988; Blalock, 1989; Besedovsky and del Rey, 1992). In addition, IL-1 is thought to exacerbate acute brain damage associated with cerebral ischemia (Touzani et al., 1999), and may play a role in acute, autoimmune destruction of myelin (Eng et al., 1996). It is also believed that IL-1 plays a role in the differentiation, proliferation, neurotransmitter release and survival of neuronal cells during development (Brenneman et al., 1992; Rothwell et al., 1996). Since IL-1β is found together with astrocytes in the brain during prenatal development, the effects of IL-1β on neurons may be mediated by the stimulation of other factors produced by astrocytes (Giulian and Lachman, 1985; Lindholm et al., 1987). Hence, the effects of IL-1β may be taken either as beneficial or detrimental to the brain. 1.3.4. Interleukin-6 IL-6 is a 26 kDa multifunctional protein produced mainly by glial cells, but also by neuronal cells. Astrocytes have been shown to be the major source of IL-6 in CNS injury and inflammation (Frei et al., 1989; Gruol and Nelson, 1997; Marz et al., 1998). The physiological role for IL-6 in the CNS is versatile. Although sometimes showing neuroprotective effects, its overexpression is generally detrimental, functioning as a mediator of inflammation, demyelization, gliosis and neurodegeneration in the brain. Therefore, IL-6 is mostly considered a proinflammatory cytokine. IL-6 has further been suggested to inhibit memory and learning as shown in a study of healthy elderly individuals where an increase in IL-6 together with a decline in cognitive ability was found (Weaver et al., 2002). This may be explained by a study showing that transgenic mice with IL-6 overexpressing astrocytes lead to decreased neurogenesis (Vallieres et al., 2002). Adult neurogenesis has been suggested to be essential for cognitive function (Gould et al., 1999; Shors et al., 2001). The age-related increase in IL-6 production has been demonstrated in a number of different studies (Prechel et al., 1996; Ye and Johnson, 1999; Ye and Johnson, 2001). The destructive potential of dysegulated IL-6 in the CNS is supported by a transgenic mouse model in which IL-6 is expressed under the control of the GFAP promoter (Campbell et al., 1993). This model leads to overexpressed IL-6, gliosis, neurodegeneration, and impaired learning, suggesting a possible role for IL-6 in neurodegenerative disorders. Furthermore, overexpression of the IL-6 gene by neurons in transgenic mice has also shown to lead to extensive gliosis (Chiang et al., 1994; Fattori et al., 1995). This suggests that the overproduction of IL-6 ultimately results in glial activation and increased inflammation in the CNS. Thus, a tight regulation of IL-6 might be important to maintain its beneficial functions and prevent its potentially destructive effects. IL-6 is known to be induced in response to inflammatory molecules like other proinflammatory cytokines, leading to a possible potentiation of ongoing inflammation (Benveniste et al., 1990; Aloisi et al., 1992; Van Wagoner et al., 1999). The transcriptional control of the IL-6 gene is somewhat complex. The IL-6 gene promoter contains binding sites for several transcription factors like NFκB, MRE, C/EBP, and AP1 that have been shown to regulate IL-6 expression (Akira et al., 1990; Libermann and Baltimore, 1990; Natsuka et al., 1992; Matsusaka et al., 1993; Chandrasekar et al., 1999; Hungness et al., 2000). Additionally, different arrangements of two or more transcription factors have been shown to be required for optimal induction of IL-6 (see section 1.4). However, NFκB appears to be the main regulator of proinflammatory INTRODUCTION 17 Inflammatory Cytokines and NFκB in Alzheimer’s Disease cytokine-induced IL-6 expression (Shimizu et al., 1990; Merola et al., 1996; Miyazawa et al., 1998; Vanden Berghe et al., 1999). IL-6 acts on target cells through a receptor complex composed of IL-6, the IL-6 receptor (IL-6R), and the signal transducing receptor gp130. This leads to the activation of various signaling pathways (for review see Taga and Kishimoto, 1997). The IL-6R can be either membrane bound or soluble (Lust et al., 1992; Mullberg et al., 1993). 1.3.5. Role of inflammatory cytokines in Alzheimer’s disease A number of cytokines, including IL-1, IL-6 and TNFα, have been found at elevated levels in AD brains (Akiyama et al., 2000; Wilson et al., 2002b). IL-1 has been shown to be overexpressed by both microglia and astrocytes in affected brain regions of AD patients (Griffin et al., 1989; Griffin et al., 1995; Sheng et al., 1997; Johnstone et al., 1999). There is also a correlation between the number of activated microglia overexpressing IL-1 and the number of senile plaques across brain regions (Sheng et al., 1998), supporting the idea that IL-1 may be of importance in the initiation and spread of Aβ plaques. Aβ has, in addition, been shown to increase the expression of several cytokines (for review see McGeer and McGeer, 2003). Further, both IL-1 and IL-6 have been shown to be able to upregulate the APP expression (Goldgaber et al., 1989; Altstiel and Sperber, 1991; Vandenabeele and Fiers, 1991; Forloni et al., 1992; Brugg et al., 1995; Griffin et al., 1998; Rogers et al., 1999) and processing (Buxbaum et al., 1992), displaying a putative direct role of these cytokines in the formation of senile plaques. It has been proposed that the overexpression of APP and Aβ deposits observed after head trauma, is a direct consequence of IL-1 and IL-6 mediated acute-phase response (Roberts et al., 1991; Gentleman et al., 1993). This consequence of induced IL-1 and IL-6, in turn, adds to the vicious cycle where processing of APP leads to more Aβ secretion, which causes further glial activation and further amplification of proinflammatory cytokines. Thereof, this cycle becomes self-propagating with sustained inflammation and maintained progression of neurodegeneration (cf., Fig. 6). A mechanism proposed to mediate cytokine-induced Aβ production is the presence of shared transcriptional elements (i.e. AP-1 and NFκB) on the promoter regions of the APP gene, as well as on the genes coding for IL-1β and IL-6 (Furutani et al., 1986; Shirakawa et al., 1993; Brugg et al., 1995). This will be discussed more in detail in the section about NFκB’s role in AD (1.4.3.). Proinflammatory cytokines can not only increase the synthesis of APP and generation of Aβ (Brugg et al., 1995; Del Bo et al., 1995; Bitting et al., 1996), but also attenuate the neurotoxic potential of the Aβ peptide (Barger et al., 1995). The induction of iNOS by Aβ in astrocytes has further been shown to require de novo IL-1β and TNFα synthesis (Akama and Van Eldik, 2000), pointing to an involvement of these proinflammatory cytokines in the Aβ-induced neurodegeneration. Moreover, impaired memory performance in transgenic mice of chronic CNS inflammation and in rats and mice treated or injected with proinflammatory cytokines has been reported (Oitzl et al., 1993; Aubert et al., 1995; Gibertini et al., 1995; Heyser et al., 1997), which strengthen the argument of the involvement of cytokines also in the behavioral characteristics for AD. The genetics of cytokines may play a role in the risk of AD. Polymorphisms of the IL-1 genes have been linked to AD, i.e., different alleles have been reported to be associated with an increased risk for AD (Grimaldi et al., 2000; Nicoll et al., 2000; 18 INTRODUCTION L. Fisher Hedley et al., 2002; Sciacca et al., 2003; Licastro et al., 2004). Individuals homozygous for IL-1α allele 2 for instance, have shown both an increased risk for AD and an earlier age of onset (Grimaldi et al., 2000). Moreover, homozygosity for both IL-1α and IL-1β allele 2 further increases the risk (Nicoll et al., 2000). These polymorphisms may possibly lead to an overexpression of the IL-1 gene, resulting in additional activation of microglia and astrocytes and increased APP production and Aβ deposition (Griffin et al., 2000). Genetic polymorphism of the IL-6 gene has also been linked to AD (Papassotiropoulos et al., 1999; Bagli et al., 2000; Pola et al., 2002; Shibata et al., 2002; Arosio et al., 2004). Not only the age of onset and the higher risk for sporadic AD has been associated with IL-6 polymorphism, but also an increased expression of IL-6 (Fishman et al., 1998; Licastro et al., 2003), indicating that increased levels of IL-6 may promote the development of AD. In addition, IL-6 has been detected in the early stages of AD, in the diffuse plaques, in brain regions yet lacking neuritic degeneration (Bauer et al., 1991; Huell et al., 1995; Hull et al., 1996a; Hull et al., 1996b). This data further points to an involvement of IL-6 in the Aβ plaque formation and progression of the disease, rather than just a consequence of neuronal degeneration. The increase in IL-6 expression in AD is, in addition, correlated with the severity of dementia (Kalman et al., 1997). Moreover, IL-6 transgenic mice show cortical pathology together with learning impairment, mimicking the pathology of AD (Chiang et al., 1994), suggesting a critical role for IL-6 in the cognitive deficits and associated inflammatory reactions documented in AD. Early induction of IL-6 in the brain of APP overexpressing transgenic mice, observed even before Aβ plaque deposits (Tehranian et al., 2001), further contribute to the notion that IL-6 appears to play a primary role in the inflammation associated with AD pathology. In addition to cytokines, the complement system, a powerful immune activator, has been shown to be activated by Aβ peptides (Rogers et al., 1992; Haga et al., 1993). The complement system has as well been shown to be overexpressed and activated in AD brain (McGeer et al., 1989; Yasojima et al., 1999), where both glial cells and neurons can produce complement proteins (Levi-Strauss and Mallat, 1987; Fischer et al., 1995; Walker et al., 1998). The expression of complement proteins in turn, leads to the activation of glial cells, thus leading to further possible damage to neurons. Moreover, IL-6 has been shown to induce the production of acute-phase proteins (Sehgal, 1990; Baumann and Gauldie, 1994), which in turn are suggested to play a role in AD by binding to Aβ and influence the aggregation of the Aβ peptide. 1.4. Nuclear Factor κB The transcription factor NFκB plays an important role as a regulator of genes involved in inflammation and immune responses, including numerous cytokines, acute-phase proteins, and immunoreceptors. The stimulatory environment, specific cell type and length/dose/type of stimuli determine whether the effect of NFκB is protective or deleterious for the host. NFκB was first discovered in 1986 as a constitutively nuclear transcription factor in mature B cells that bound to an element in the immunoglobulin κ light-chain enhancer, and thereof derived its name (i.e. nuclear factor κB) (Sen and Baltimore, 1986). As in other organ systems, NFκB is present in essentially all cell types INTRODUCTION 19 Inflammatory Cytokines and NFκB in Alzheimer’s Disease in the nervous system, including neurons, astrocytes, and microglia (O'Neill and Kaltschmidt, 1997). NFκB consists of a complex of two subunits and is found in the cytoplasm in an inactive form bound to an inhibitory protein termed IκB. IκB presumably interacts with NFκB in or around the nuclear localization sequence, thus keeping it from translocating into the nucleus (Beg et al., 1992; Ganchi et al., 1992; Henkel et al., 1992). In addition, IκB exhibits a nuclear export signal, which helps keeping the inactive form of NFκB in the cytoplasm (Arenzana-Seisdedos et al., 1995; Johnson et al., 1999). The key steps leading to NFκB activation and nuclear translocation after external stimuli involves phosphorylation of two specific serine residues of IκB by IκB kinase (IKK). This result in ubiquitination followed by degradation of IκB by the proteosome, allowing NFκB to translocate to the nucleus (see Fig. 5). Inside the nucleus, NFκB binds to the κB consensus sequence in promoters of target genes, generally leading to an increase in the expression of the gene (Baeuerle and Henkel, 1994; Siebenlist et al., 1994; Karin, 1999). IκB is a member of a larger family of inhibitory molecules. The best characterized IκB is ΙκBα, mainly because it was the first member of this family to be cloned (Davis et al., 1991; Haskill et al., 1991). NFκB influences the expression of a complex array of genes in the nervous system, most of which are important in cellular responses to inflammation, in particular IL-1, IL-6, TNFα and interferon-γ (IFN-γ). Many of these gene products can, in turn, directly activate the NFκB pathway to establish a positive auto-regulatory loop that can add to and intensify the duration of the inflammatory response. Moreover, acute-phase proteins and iNOS have been shown to be regulated by NFκB in response to IL-1 and IL6 (Grilli et al., 1993; Kopp and Ghosh, 1995; Tomita et al., 1998). In addition to immune and inflammatory related genes, NFκB also regulates genes involved in the control of cell growth and apoptosis (Pahl, 1999). As a negative feed-back loop, controlling the activity and quantity of activated NFκB, NFκB also cause the transcriptional activation of the IκBα gene (Ten et al., 1992; Le Bail et al., 1993). The re-synthesized IκB then interacts with NFκB and probably relocates it from the nucleus back to the cytoplasm by a nuclear export sequence (Arenzana-Seisdedos et al., 1995; Johnson et al., 1999). Many of the immunoregulatory genes that are activated by NFκB are also subject to control by other transcription factors. Moreover, interactions between NFκB and other transcription factors may influence the ability of NFκB to regulate the expression of a specific gene. An example of interaction between NFκB and other transcription factors to activate a particular gene after a particular stimulus was demonstrated by Thanos and Maniatis. They showed that TNF-induced activation of the IFN-β gene requires a multicomponent complex including three distinct transcription factors, one of them being NFκB (Thanos and Maniatis, 1995). Another example of synergistic activation of transcription due to interactions of NFκB and other transcription factors is found within the human immunodeficiency virus 1 (HIV-1), where activation is dependent on both NFκB and Sp1 binding to the promoter (Perkins et al., 1993; Perkins et al., 1994). NFκB can also interact with AP-1 for enhanced DNA binding and transcription of specific target genes (Stein et al., 1993). Transcriptional synergy between NFκB and C/EBP has also been demonstrated (LeClair et al., 1992; Li and Liao, 1992; Betts et al., 1993; Matsusaka et al., 1993; Stein and Baldwin, 1993; Diehl and Hannink, 1994; Dunn et al., 1994; Shimizu and Yamamoto, 1994; Ray et al., 1995; Lee et al., 1996; Kinugawa et al., 20 INTRODUCTION L. Fisher 1997; Plevy et al., 1997; Xia et al., 1997). For example, the inhibition of the IL-6 gene by the estrogen receptor has been shown to be mediated through both the NFκB and C/EBP sites in the IL-6 gene (Ray et al., 1994; Stein and Yang, 1995). Figure 5. IL-1β-induced NFκB activation. IL-1β binds to its receptor, IL-1RI, which leads to the formation of the IL-1R and IL-1RAcP complex. The adaptor protein MyD88 is recruited, which in turn leads to the interaction of the IL-1 receptor-associated kinase (IRAK). IRAK then leaves the complex and interact with TRAF-6. This leads to the activation of IKK with resultant IκB degradation and thus NFκB activation. There is a range of different NFκB inducers, including cytokines, growth factors, oxidative stress, along with bacteria and viruses and their gene products (Pahl, 1999). In light of the great diversity of stimuli that can activate NFκB, it is clear that several upstream pathways can lead to IκB phosphorylation and degradation. Although all these pathways are not clearified, one of the most studied NFκB activation pathways, induced by IL-1β, is well characterized (Fig. 5) (Burns et al., 1998; Mochida et al., 2000). 1.4.1. The Rel family of proteins The NFκB proteins are part of the NFκB/Rel protein family, which in mammals consists of five members characterized by the Rel homology domain (RHD). The RHD domain contains the nuclear localization signal and is accountable for the dimerization, IκB interaction, and DNA binding of NFκB. The members of the NFκB/Rel family; p65 INTRODUCTION 21 Inflammatory Cytokines and NFκB in Alzheimer’s Disease (RelA), RelB, c-Rel, p50, and p52 form different NFκB homo- or heterodimer complexes, with p50/p65 being the most frequent one (Siebenlist et al., 1994; Baldwin, 1996). An active NFκB dimer requires either the p65, RelB, or c-Rel subunit to induce gene expression, since they are the only members containing C-terminal transcriptional activation domains crucial for their ability to activate gene expression. Each of the various NFκB dimers may exhibit distinct properties, like preferable binding sites or cell type specific expression. The favoured binding site for different NFκB dimers has been demonstrated to differ (Kunsch et al., 1992). The p65/c-Rel dimer for instance, has been found to bind to a sequence different from the binding sequence of p50/p65 (Parry and Mackman, 1994). Studies in vivo and in vitro also indicate that different NFκB dimers have different transcriptional activation properties (Schmid et al., 1991; Lin et al., 1995). Different NFκB complexes have further been shown to be expressed in different cell types or at different developmental stages (Bakalkin et al., 1993; Perkins, 1997). In addition, a neuronal κB factor, distinct from the common NFκB subunits, has been described (Moerman et al., 1999). Knockout mice of the individual members of the NFκB/Rel family further reveal distinct and unique roles for each and every one of them (for review see Ghosh et al., 1998). The different NFκB subunits may be regulated by different stimuli. The selectivity of DNA binding by specific NFκB complexes, in addition gives the potential to regulate the expression of different genes, which is crucial for the role of NFκB in both cell survival and apoptosis. However, we still have little knowledge about how specific NFκB complexes contribute to a given physiological response. 1.4.2. NFκ κB in neurons and glial cells There are many lines of evidence for an anti-apoptotic role for activated NFκB in neurons (for review see Mattson et al., 2000). It has been suggested that NFκB promotes cell survival through stabilizing intracellular calcium levels (Cheng et al., 1994; Schneider et al., 1999). It has also been suggested that activated NFκB may increase neuronal survival by activating several anti-apoptotic proteins. NFκB may control apoptosis during the development of the nervous system, as suggested by several studies (Maggirwar et al., 1998; Middleton et al., 2000). It may also play a role in synaptic plasticity (Furukawa and Mattson, 1998; Albensi and Mattson, 2000). In contrast to this, activated neuronal NFκB have also been demonstrated to promote cell death (Draczynska-Lusiak et al., 1998; Luo et al., 1999). Suppression of NFκB activity has, in addition, been shown to be neuroprotective both in vitro (Grilli et al., 1996b; Post et al., 1998) and in vivo (Qin et al., 1998; Schneider et al., 1999). Based on these facts, it has been suggested that the crucial role for NFκB in the survival or death of neurons is dependent on the stimulus and activation kinetics of NFκB (Lipton, 1997; Kaltschmidt et al., 2000; Pizzi et al., 2002). In neurons, NFκB exists both in an inducible form and a constitutively active form (Kaltschmidt et al., 1993; Kaltschmidt et al., 1994b). The fact that constitutive NFκB activity is required for neuronal survival (Bhakar et al., 2002) may, due to the dual faces of NFκB, point to the involvement of inducible NFκB in neurodegeneration. One also has to consider that the contradictory effects of NFκB may be explained by the involvement of different NFκB complexes that may affect the cell in different ways. 22 INTRODUCTION L. Fisher However, the complexity and conflicting effects of NFκB still remains unclear and a hot matter of debate. In glial cells, NFκB regulates the cytokine cascades (Sparacio et al., 1992; Moynagh et al., 1993; Kaltschmidt et al., 1994a; papers II and III), suggesting that it may indirectly lead to apoptosis of neurons by promoting production of cytotoxic agents such as NO and cytokines (Qin et al., 1998). Activation of NFκB in astrocytes results in increased expression of iNOS and increased NO production as well as IL-6 and other proinflammatory cytokines. Activation of NFκB in microglia promotes ischemic neuronal degeneration, whereas in neurons, activation of NFκB probably increases their survival. Hence, the pro- or anti-apoptotic action of NFκB probably depends on, not only activating stimuli but also the specific cell type. In general it appears that the activation of NFκB in neurons protects them against degeneration, while activation of NFκB in glial cells promotes neuronal cell death. 1.4.3. Role of NFκ κB in Alzheimer’s disease Studies of postmortem brain tissue from patients with AD have revealed increased NFκB activity in neurons as well as astrocytes in the immediate vicinity of amyloid plaques (Terai et al., 1996; Boissiere et al., 1997; Kaltschmidt et al., 1997; Kitamura et al., 1997; O'Neill and Kaltschmidt, 1997). Aβ has further been demonstrated to activate NFκB and subsequent gene expression through κB sites (Behl et al., 1994; Barger et al., 1995; Akama et al., 1998; Bales et al., 1998; Guo et al., 1998; Dodel et al., 1999; Kim et al., 2003; papers II and III), suggesting a role for Aβ in the NFκB activation observed in AD. The activation of NFκB by Aβ has been shown to lead to the induction of M-CSF, which will contribute to the ongoing inflammation detected in AD brains (Du Yan et al., 1997). In addition, the 5´ regulatory region of the gene encoding APP contains κBbinding sites (Grilli et al., 1995; Grilli et al., 1996a). In fact, NFκB activation can increase APP production with subsequent increased Aβ1-42 generation (Tomita et al., 2000b). Since NFκB is both being activated by and induce the expression of cytokines and APP, this gives rise to the possibility for NFκB activation to remain high and cause further damage in AD (see Fig. 6). Thus, a dysregulation of the NFκB pathway may be involved not only in the inflammation occurring in the AD brain, but also in promoting the generation of Aβ. NFκB may even take part in mediating Aβ-induced neurotoxicity. A recent study demonstrates a pivatol role for microglial NFκB signaling in mediating Aβ toxicity (Chen et al., 2005). They show that inhibiting Aβ-induced NFκB activation in microglia had strong neuroprotective effects, supporting the idea of a critical role of NFκB in AD. In addition to inflammatory cytokines, Aβ-induced iNOS expression and ROS production have been suggested to be mediated through an NFκB-dependent pathway (Akama et al., 1998; Akama and Van Eldik, 2000). The occurrence of free radicals like ROS and NO can cause damage to crucial compartments of the cell, resulting in oxidative stress, ultimately leading to cell death. Oxidative stress has been proposed to be one of the key factors contributing to the neurodegeneration observed in AD. The lesions present in the brains of AD patients are very similar to lesions attacked by free radicals i.e. DNA damage, protein oxidation, lipid peroxidation, and glycooxidation resulting in the formation of AGE (Subbarao et al., 1990; Mecocci et al., 1994; Palmer and Burns, INTRODUCTION 23 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 1994; Yan et al., 1994; Hensley et al., 1995; Marcus et al., 1998; Koppal et al., 1999; Pratico and Delanty, 2000; Butterfield et al., 2002). In addition, ROS has been shown to accumulate in the aging brain (Benzi and Moretti, 1995), causing damage to cells. The fact that age is the number one risk factor for sporadic AD offers more support to this theory, that oxidative stress is indeed involved in the progression of the disease. Several studies have shown that the aggregated Aβ peptide induces oxidative events (Butterfield et al., 1994; Hensley et al., 1994; Harris et al., 1995b; Behl, 1999), which have further been linked to Aβ toxicity (Xie et al., 2002). Reports showing that antioxidant therapies can be beneficial in the treatment of AD patients (Le Bars et al., 1997; Sano et al., 1997; Gutzmann and Hadler, 1998) substantiate the involvement of oxidative stress in AD. Additionally, antioxidants, including vitamin E, have been shown to prevent Aβ toxicity (Behl et al., 1992; Behl et al., 1994; Goodman et al., 1994; Behl et al., 1997; Sano et al., 1997; Butterfield et al., 1999; Bisaglia et al., 2004). The fact that these antioxidants also have been shown to inhibit NFκB activation and protect the cells from oxidative stress (Grilli et al., 1996b; Ayasolla et al., 2004), suggests a role for NFκB in the oxidative stress-mediated cell death observed in AD. However, other studies provide oppose results, showing that NFκB activity mediates protection to oxidative stress (Lezoualc'h et al., 1998; Kaltschmidt et al., 2002). Figure 6. Illustration of how activation of NFκB by Aβ or proinflammatory cytokines can lead to more APP and cytokine expression, oxidative stress and M-CSF production, which in turn will set up for a vicious cycle where more activated NFκB with subsequent neurotoxic agents ultimately will lead to neuronal death. 24 INTRODUCTION L. Fisher Also in regard to AD and Aβ-induced NFκB activation, the activation of NFκB is a double-edged sword. NFκB activation has been shown to inhibit the neurotoxic action of Aβ (Barger et al., 1995; Mattson et al., 1997). In addition, the suggested protective role for NFκB in neurons is supported by reports proposing that the protective role for sAPPα can be attributed the activation of NFκB in neurons (Barger and Mattson, 1996; Guo et al., 1998). On the contrary, decreased levels of NFκB activation in neuronal cells have also been associated with protection against Aβ toxicity (Bisaglia et al., 2002). Another study has also shown that inhibiting NFκB activation reduces Aβ-induced neuronal cell death (Song et al., 2004). In yet another study, a correlation between Aβinduced neuronal DNA damage and neuronal NFκB activation was found in cerebral regions of AD patients (Garcia-Ospina et al., 2003), suggesting a pro-apoptotic role for NFκB. 1.5. Interference with gene expression Methods to control and interfere with endogenous gene expression are important both for functional genomics and for potential therapeutic applications. In eukaryotes, gene expression can be controlled by a variety of mechanisms that range from those that prevent transcription to those that prevent activation after the protein has been produced. The different mechanisms can be placed into one of the following three categories: transcriptional, translational, and posttranslational. The interference of gene expression at a transcriptional level, that is, to prevent or activate transcription of a DNA sequence into an RNA transcript, is the most important and widely used strategy. Except for direct interference with the gene, e.g. gene knockout, these mechanisms often involve the inhibition or activation of transcription factors, which in turn reduce or enhance the activity of RNA polymerase at a given promoter on a specific gene. The activation of transcription factors is regulated by signals produced from other molecules, like for instance kinases. Phosphorylation and dephosphorylation are key steps in the modification of many transcription factors. Several clinically used drugs target this aspect by modulating the phosphorylation state of one or more transcription factors (Pierce et al., 1996; Shibasaki et al., 1996; Beals et al., 1997). Interactions of transcription factors with other proteins are also central to the regulation of their activity. Such interactions may as well be targeted to control different transcription factors. Moreover, the binding of the transcription factor to the target gene can be hindered. This may be accomplished by transcription factor decoys which will be discussed more in detail in section 1.5.2. In addition, steroid hormones and protease inhibitors have been shown to interact directly with transcription factors to inhibit their action (Singh and Aggarwal, 1995a; Rossi et al., 2000; Davies et al., 2004; Iwasaki et al., 2004). It should be mentioned that, since many different genes and many different types of cells share the same transcription factors, the approach of controlling transcription factors is useful when it is desirable to switch on or off an entire pool of genes, involved in for example inflammation. In such a case, it would clearly be more effective to target the transcription factor itself that controls these processes, rather than each of the proteins encoded by its target genes. In many situations however, it is preferable to block single genes, which is usually done by posttranscriptional control. INTRODUCTION 25 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Controlling gene expression on a translational level means regulating mRNA after it has been produced to prevent the synthesis of a protein. Antisense technology is a tool used to interfere with gene expression mainly at this level. The principle behind it is that an antisense nucleic acid sequence base pairs with its complementary mRNA strand, and prevents it from being translated into a protein. The exact mechanism by which translation is blocked is still unknown. It may be due to that the complimentary nucleic acid sequence, after hybridization to mRNA, interferes with ribosomal activity or that the rate by which the dsRNA is degraded is accelerated. The complimentary nucleic acid sequence can be either a synthetic oligonucleotide, often oligodeoxyribonucleotides (ODNs) of less than 30 nucleotides, or longer antisense RNA sequences (Sczakiel, 1997). ODNs are usually used since they are more stable than RNAs in cellular environments. Another mechanism for gene interference using synthetic dsRNA has been developed in the past few years (for review see Bosher and Labouesse, 2000). The ability of a dsRNA to suppress the expression of a gene corresponding to its own sequence is called RNA interference (RNAi) or posttranscriptional gene silencing. RNAi works through the introduction of gene-specific dsRNA into a cell, causing the homologous mRNA to be degraded (Cottrell and Doering, 2003). Since long dsRNA have been shown to trigger a non-specific shutdown of all protein synthesis in mammalian cells (Gil and Esteban, 2000), short interfering RNA (siRNA) with less than 23 nucleotides is usually used (Elbashir et al., 2001; Brummelkamp et al., 2002; Lee et al., 2002; Sui et al., 2002; Scherr et al., 2003). Finally, interference with gene expression can be done at a posttranslational level, after the protein has been produced. Many proteins, like IL-1β, are produced in an inactive form and must be further processed in order to become active. Other proteins may require activation by being combined with some other molecule, or by being freed from another molecule, for example by phosphorylation. 1.5.1. PNA as antisense instrument In the past several years, alternatives to oligonucleotides have been developed, including peptide nucleic acids (PNAs), to block the transcriptional and translational processes inside cells. PNA is an uncharged DNA mimic with a pseudopeptide backbone composed of N-(2-aminoethyl)glycine units (Nielsen et al., 1991). PNA molecules are extremely stable in biological systems as they are resistant to both nuclease and protease degradation (Demidov et al., 1994). In addition, charged antisense oligonucleotides may possess non-specific effects (Branch, 1998), which are minimized with the use of these neutral PNA molecules. PNAs hybridize to complementary sequences of single stranded DNA and RNA with high affinity and specificity, forming Watson-Crick double helices. Since PNAs are resistant to proteases and nucleases, bind independently of salt concentration to their complementary nucleic acids, have higher affinity for nucleic acids, and are more gene specific than DNA/DNA duplexes (Egholm et al., 1993; Nielsen et al., 1993; Demidov et al., 1994; Corey, 1997; Jensen et al., 1997; Giesen et al., 1998), they can serve as high-quality antisense and antigene agents. In addition, PNAs are fairly easy to synthesize and modify. PNAs have been shown to effectively downregulate individual genes, both in vitro (Hanvey et al., 1992; Bonham et al., 1995; Vickers et al., 1995; Knudsen and Nielsen, 1996; Kilk et al., 2004, paper IV) and in vivo (Tyler et al., 1998; 26 INTRODUCTION L. Fisher McMahon et al., 2002b; Boules et al., 2004; Suzuki et al., 2004). However, being a large hydrophilic molecule, PNA does not cross lipid membranes easily. 1.5.2. Transcription factor decoy The basic theory behind the decoy approach involves overflowing the cell with competing synthetic, transcription factor-specific consensus sequences. These synthetic decoys, if delivered into the cell, will compete for binding of the transcription factor with consensus sequences in the authentic binding elements of the promoter regions of target genes and interfere with gene regulation. These decoys have been reported to completely block transcription factor functions. Evidently, they represent powerful research tools for studying gene regulation both in vitro and also more recently in vivo (for review see Morishita et al., 1998; Mann and Dzau, 2000). Using the decoy approach to regulate transcription factors may be beneficial since it does not affect the actual production of the target protein, but rather its capacity to bind to the regulatory DNA element. The decoy approach against NFκB has been proposed as a useful tool to alter NFκB dependent gene expression. It has previously been shown that the decoys can downregulate NFκB activation and subsequent gene expression both in vitro (Mattson et al., 1997; Schmedtje et al., 1997; Khaled et al., 1998; Tomita et al., 1998; Tomita et al., 2000a; Ye and Johnson, 2001; Gao et al., 2002) and in vivo (Morishita et al., 1997; Kawamura et al., 1999; Tomita et al., 2000c; Kawamura et al., 2001). However, the uptake of DNA into mammalian cells is not very efficient, and therefore, as shown by those studies, high concentrations and long incubation times have been necessary to achieve significant effects. 1.6. Cellular uptake of PNA and oligonucleotides Although unmodified PNAs and oligonucleotides have been shown to be able to affect cellular processes (Good and Nielsen, 1998; Sei et al., 2000; paper IV), they have demonstrated poor cellular uptake in living cells (Wittung et al., 1995; Gray et al., 1997). The long incubation times and high concentrations needed may further induce toxic stress to cells. Different cellular delivering systems for PNAs as well as oligonucleotides have been employed during the last few years; microinjection, transfection, electroporation, and peptide-conjugation to mention a few (for review see Koppelhus and Nielsen, 2003). 1.6.1. Cell-penetrating peptides In recent years, the use of peptides as carriers for cellular delivery of PNAs and oligonucleotides has gained much attention. These transporting molecules referred to as cell-penetrating peptides (CPPs), have been reported to greatly improve the transport of not only oligonucleotides and PNAs, but also peptides, proteins, plasmids, and viruses across biological membranes (Aldrian-Herrada et al., 1998; Pooga et al., 1998b; Gratton et al., 2003; Howl et al., 2003; Ignatovich et al., 2003; Begley et al., 2004; Caron et al., 2004; Tessier et al., 2004; paper I and III). CPPs are short peptide sequences, usually no longer than 30 amino acids in length. They have been suggested to be more efficient in their cellular internalization if the peptide sequence includes more positive charges and INTRODUCTION 27 Inflammatory Cytokines and NFκB in Alzheimer’s Disease bulky hydrophobic side-chain amino acids (Derossi et al., 1998; Mitchell et al., 2000; Wender et al., 2000; Futaki et al., 2001). Still, the primary structure of CPPs differs tremendously, proposing that the means by which they cross the plasma membrane may vary. CPPs have been proposed to penetrate cells in a receptor-independent manner (Derossi et al., 1994; Derossi et al., 1996; Derossi et al., 1998). The cell penetration by these peptides has also, by some research groups, been considered to be an energyindependent mechanism, hence mediated by a non-endocytotic pathway (Derossi et al., 1998; Pooga et al., 1998a; Silhol et al., 2002; Suzuki et al., 2002; Lindgren et al., 2004). However, lately, new research results have demonstrated that this is probably not completely true. It has been shown that the apparent cellular distribution of CPPs inside the cell observed in fixed cells may be due to artifacts i.e. the peptides may relocate upon fixation. The consequence of this and the possible mechanism by which CPPs penetrate the plasma membrane will further be discussed in section 4.1. 28 INTRODUCTION L. Fisher 2. AIMS OF THE STUDY Knowing that inflammation is a probable contributor to the progression of Alzheimer’s disease, blocking the transcription factor involved in the synthesis of many inflammatory genes may be of great therapeutical value in the search for a treatment for this disease. Hence, the general aim of this thesis was to investigate the role of NFκB in the βamyloid-induced cytokine expression in primary glial cells, and to study the possibility to block the induced cytokine expression by blocking NFκB. The specific aims were to: Investigate whether a cell-penetrating transport peptide could be used to facilitate the translocation of an NFκB decoy across the plasma membrane (paper I). Investigate the effect of IL-1β and Aβ on NFκB activation in primary glial cells (papers II and III). Investigate the effects of IL-1β and Aβ alone or combined on cytokine expression in primary glial cells (papers II and III). Investigate the possibility to block the NFκB binding activity in primary glial cells using an NFκB decoy together with a cell-penetrating transport peptide (paper III). Investigate whether blocking NFκB had any effect on the IL-6 expression in primary glial cells (paper III). Investigate if it is possible to downregulate APP in primary neuronal and glial cells using PNA antisense (paper IV). AIMS OF THE STUDY 29 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 3. METHODOLOGICAL CONSIDERATIONS The methods used in this thesis are described in detail in each paper. In this section some theoretical and methodological aspects will be discussed. 3.1. Cell cultures 3.1.1. Rinm5F cell line In paper I, rat Rinm5F insulinoma cells were used as a model system for investigating the cellular uptake of the NFκB decoy bound to a cell-penetrating peptide via PNA. The Rinm5F cell line is a homogenous population of immortalized β-cells from the Islet of Langerhans area of the pancreas. These cells express a high density of IL-1 receptors (Sandler et al., 1989; Corbett et al., 1992; Bristulf et al., 1994), and are therefore often used when investigating the effects of IL-1. Since we used IL-1β as an activator of NFκB, these cells were a natural choice for this study. 3.1.2. Primary mixed glial cell cultures In papers II-IV, primary mixed glial cultures containing both astrocytes and microglia from newborn Sprague Dawley rats were used. Astrocytes from animals less than 24 h of age show a high proliferative rate, and the cultures used contained about 90% astrocytes and 10% microglia. To avoid a mixed cell culture containing mostly fibroblasts, which have a higher proliferative rate compared to astrocytes, the fibroblast containing meningal membranes were removed. Astrocytes, together with microglia, are the primary source of cytokine production in the brain during inflammatory conditions. Therefore, we chose to work with mixed cultures in paper II and III, where the effects of Aβ and IL-1β on cytokine expression and secretion were investigated. These mixed glial cultures were also used in paper IV were we wanted to compare the downregulation of APP and the cellular uptake of PNA in primary cultured neurons and astrocytes. It is also possible to use pure astrocytes or microglia cultures. This may be an advantage if the aim is to determine the cellular source of a specific cytokine. On the other hand, mixed glial cultures resemble the brain more, and the interaction between astrocytes and microglia can be studied. It has also been shown that both contacts and products secreted by astrocytes play a part in generating the adult phenotype of microglia in culture (Sievers et al., 1994). As an alternative to the primary glial cultures, there are many cell lines available with many characteristics comparable to the primary glial cultures that could be used. The benefits of using cell lines are the ease of maintenance and also to ensure purity of the cultures. However, the advantage of using primary cell cultures is that they are known to maintain most of the biological properties of the cells in vivo. Glioma and astrocytoma cell lines, on the other hand, are often isolated from tumor tissue of the CNS, and differ from normal cells in that they are immortal and proliferative. 3.1.3. Primary neuronal cell cultures In paper IV, primary cultures of cerebellar granule cells (CGCs) from Sprague Dawley rats were used to study the downregulation of APP and the cellular uptake of PNA. CGCs represent the largest homogeneous neuronal population of mammalian brain (Thangnipon 30 METHODOLOGICAL CONSIDERATIONS L. Fisher et al., 1983; Zirrgiebel et al., 1995). Due to their postnatal generation and continuing proliferation and differentiation after birth, the CGCs are a common model of choice for the study of cellular and molecular mechanisms in for instance neurodegeneration and neuroprotection. Accordingly, we chose these cells to investigate the uptake and antisense effect of PNA directed against APP mRNA, compared to primary astrocyte cell cultures. 3.2. Design of PNA and transport peptide-PNA constructs In papers I and III, PNA oligomers together with a CPP were used to increase the cellular uptake of an NFκB decoy. This CPP-PNA construct was used as a cargo delivery system, where the PNA sequence is complementary to the single-stranded protruding 3´-terminal end of the NFκB oligonucleotide (Fig. 2, paper I). The CPPs transportan (TP) and transportan 10 (TP10) were chosen as they have shown good cellular cargo transportation capabilities. TP is a 27 amino acids long CPP (Table 2). It is a chimeric peptide, constructed from the neuropeptide galanin, and the wasp venom peptide mastoparan (galanin (1-12)-Lys-mastoparan (1-14) amide) (Pooga et al., 1998a). It has previously been used successfully to deliver PNA, peptides, and proteins into cells (Pooga et al., 1998b; Pooga et al., 2001; Ostenson et al., 2002). TP10 is a short analogue of TP, lacking the first six N-terminal amino acids (Soomets et al., 2000). The TP peptides have been shown to be more stable and to cross the epithelial cell layer more efficiently than, for example, the CPP penetratin (Lindgren et al., 2004). Further, these peptides, especially the shorter analogue, have been shown to demonstrate minor or no toxicity as shown by membrane disturbance assays, measurement of GTPase activity, and LDH release (Pooga et al., 1998a; Soomets et al., 2000; Lindgren et al., 2004; Nekhotiaeva et al., 2004; Saar et al., 2005). A disulfide bond was chosen to link the transport peptide with the PNA. The disulfide bond is ideal as a link between the peptides and PNA, due to the fact that, once inside the cell, the disulfide bond will be reduced, releasing the cargo molecule from the transport peptide. The cargo molecule, here NFκB decoy, is then no longer attached to the transport peptide which otherwise possibly can disturb the effect of the decoy when inside the cell. Table 2. CPP sequences examined in this thesis. The sequences are shown as one-letter code for the amino acids. Name Transportan Transportan10 Sequence GWTLNSAGYLLGKINLKALAALAKKIL-NH2 AGYLLGKINLKALAALAKKIL-NH2 In paper IV, a biotinylated PNA oligomer directed to the initiator codon region of the APP mRNA (Table 3) was used to study the cellular uptake and antisense effect of otherwise unmodified PNA. Biotin can easily be attached to the free amino groups of the PNA using solid phase peptide synthesis. METHODOLOGICAL CONSIDERATIONS 31 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Table 3. DNA sequences of different PNAs and oligonucleotides examined in this thesis. Name PNA(9) PNA(6) PNA(APP) NFκB Decoy NFκB Scramble Sequence ACACAAGGA ACACAA CTGGGCAGCATCGTG 5´-AGTTGAGGGGACTTTCCCAGGC-3´ 5´-GCCTGTCGACGACGACTCAACT-3´ 3.3. Cell treatments In paper II, the full length Aβ peptide, Aβ1-42, found in the senile plaques characteristic of AD brain tissue, was used. In view of the fact that Aβ deposits are widely believed to contribute not only to the progressive neurodegeneration observed in AD, but also to the early inflammation observed, we used this peptide to investigate its effect on cytokine expression in primary mixed glial cultures from rat. Since it has been shown that it is the aggregated form of the Aβ peptide that forms the extracellular senile plaques, the peptide was incubated at 37°C for 72 h to allow aggregation. Though debated, many in vitro studies have also shown that it is only aggregated forms of Aβ peptides that can reproducibly exert toxic effects on neurons and activate glial cells (Pike et al., 1994; Zhang et al., 1994; Casal et al., 2002). Using Congo red staining, we estimated the time needed for each of the Aβ peptides to form aggregates. We found that the Aβ1-42 peptide formed aggregates within 72 h in culture medium at 37°C, and Aβ25-35 within 30 min. In order to minimize variations in the aggregated form of the different peptides used in different experiments, all peptides were initially dissolved in hexafluoroisopropanol, known to disaggregate amyloid fibrils. The small neurotoxic fragment of the Aβ peptide, Aβ25-35, was used in papers II and III. The short Aβ25-35 peptide is a synthetic derivative of Aβ not found in real life. It is often used as a model peptide for the full length Aβ peptide, as it displays similar effects as Aβ1-42, but is easier to synthesize and less expensive. Aβ25-35 is the shortest Aβ fragment that exhibits large β-sheet fibrils and the same retained bioactive properties as the full length Aβ peptides (Pike et al., 1993; Shearman et al., 1994; Terzi et al., 1994; Pike et al., 1995b). A correlation between the aggregation state of the Aβ peptides and the toxicity of the peptides (Pike et al., 1993) may explain the similar neurotoxic effects observed from Aβ25-35 compared to full length Aβ (see also section 1.1.5). In papers I through III, the proinflammatory cytokine IL-1β was used, as it is known to activate NFκB. In addition, since IL-1β has also been proposed to play a role in neurodegenerative processes where it in turn can induce the expression of other cytokines, it was used in paper II and III to investigate the regulation of IL-6 expression in rat primary mixed glial cultures. Here we also investigated the effect of IL-1β together with either the Aβ1-42 peptide or the Aβ25-35 peptide. 32 METHODOLOGICAL CONSIDERATIONS L. Fisher 3.4. Analysis of specific protein-DNA interactions The interaction of proteins and DNA is central to the control of many cellular processes including replication, transcription and regulation of gene expression. Therefore, it is of great importance to be able to study these interactions. Defining specific interactions can help in the study of gene regulation and drug discovery. There are several methods used to study specific protein-DNA interactions, including foot printing, filter binding, X-ray crystallography and EMSA (electrophoretic mobility shift assay). Foot printing can be used to analyze the binding site in the DNA to which the protein binds to. The filterbinding assay is a quick and easy method to investigate protein-DNA interactions, but this method cannot reveal multiple complexes. The X-ray crystallography technique can give precise structural information of the interaction, but this method is extremely demanding and time consuming. The EMSA technique is based on the fact that proteinDNA complexes migrate more slowly than free DNA fragments when separated by gel electrophoresis. An advantage of studying protein-DNA interactions by electrophoresis is that it is possible to separate complexes not only with different molecular weight, but also with different conformation. Another major advantage for many applications is that the source of the DNA-binding protein may be a nuclear or whole cell extract rather than a purified preparation. 3.4.1. Electrophoretic mobility shift assay In papers I-III, EMSA was used to monitor the activation and inhibition of the transcription factor NFκB. It has been shown to be the simplest and most reliable assay to demonstrate NFκB activation (Muller et al., 1997). EMSA can be used qualitatively to identify sequence-specific DNA-binding proteins (such as transcription factors) in crude lysates. The protein containing extract is mixed with labeled DNA, often in the form of synthetic oligonucleotides, in a binding reaction to form a complex. Conventionally, DNA is radio labeled with 32P by 5´ end labeling using γ-32P-ATP and T4 polynucleotide kinase. Nonspecific competitor DNA such as poly(dI•dC) or poly(dA•dT) is included in the binding reaction to minimize the binding of nonspecific proteins to the labeled target DNA. Since some proteins in the crude lysate will bind to any general DNA sequence, these repetitive polymers will provide an excess of nonspecific sites that these proteins can bind to. A specific unlabeled competitor of the same sequence as the target DNA can also be added in excess to the binding reaction to compete away the specific interaction. Thus, any detectable specific band should be abolished by the presence of excess unlabeled specific competitor. A second control can be used to identify the specific band; a so-called supershift assay can be performed. Here an antibody that recognizes the protein of interest is added to the binding reaction mixture. If the complex shifts further up in the gel, creating a supershift, this is evidence that the protein was present in the initial complex. The binding of the antibody to the protein may also be observed as a decrease of the protein-DNA complex band intensity. Following the binding reaction, the protein-DNA complex is separated from free DNA by non-denaturing polyacrylamide or agarose gel elecrophoresis (Fig. 7). The gel percentage used depends on the size of the target DNA and the size, number and charge of the protein(s) bound to it. An important aspect of electrophoresis that allows a band shift to work appropriately is the cage effect. Once a complex enters the gel, the gel METHODOLOGICAL CONSIDERATIONS 33 Inflammatory Cytokines and NFκB in Alzheimer’s Disease matrix provides a cage such that if the complex subsequently dissociates, the components tend to reassociate rather than diffuse away from each other. Therefore, even labile complexes can often be resolved by this method. Finally, the individual protein-DNA complexes can be visualized as distinct bands within the gel using radioisotopic detection or chemiluminescence. Figure 7. Schematic illustration of the general EMSA approach. Nuclear proteins are allowed to bind to labeled ds oligonucleotides mimicking the response element of a specific gene. The protein-DNA complex can then be separated from unbound oligonucleotide and antibody-bound protein-DNA complex. 34 METHODOLOGICAL CONSIDERATIONS L. Fisher 3.5. Analysis of mRNA expression The quantification of mRNA in a cell can be useful to detect modest changes in different cells after, for example, different treatments. When the protein levels are too low to detect, the mRNA can provide an indication of the corresponding protein level. However, changes of mRNA quantity are not always exactly correlated to changes of protein quantity. The decomposition rates of mRNA and protein are different, and proteins can undergo several modifications within a cell. Nevertheless, changes in mRNA levels give us information on the cellular response on a transcriptional level. It enables us for instance to investigate which factors induce the expression of a particular gene and the time scale for this induction. 3.5.1. RT-PCR analysis RT-PCR (reverse transcription-polymerase chain reaction) is a technique for measuring changes in gene expression at the mRNA level. In papers I-III, relative RT-PCR analysis was used to detect differences in cytokine mRNA levels after various treatments. RNA is isolated from tissue or cells followed by copying the RNA template into a complementary DNA (cDNA) transcript using primers and reverse transcriptase. In our studies, we used a random primer since an rRNA was used as internal standard. Otherwise, an oligo dT could be used to reverse transcribe only mRNA. The cDNA is thereafter amplified exponentially using PCR. This step requires knowledge of sequences that flank the region to be amplified. Oligonucleotides that are complementary to these sequences are then used as primers in the DNA polymerase catalyzed PCR (Fig. 8). The PCR product is finally detected, most commonly by agarose gel electrophoresis and ethidium bromide staining. Since the cDNA can be amplified into more than a million copies, this technique is very sensitive to DNA contamination and much care must be taken to make sure that the samples are free from genomic DNA. However, this problem is usually solved by designing primers of which the amplified sequence includes one intron-splice site in the genomic DNA, allowing the amplification products derived from cDNA and contaminating genomic DNA to easily be distinguished. Furthermore, this also makes RT-PCR the most sensitive technique for detection and quantification of mRNA expression available today. Compared to other frequently used techniques for quantifying mRNA levels like Northern blot analysis and ribonuclease protection assay (RPA), RT-PCR can be used to quantify mRNA levels from much smaller samples. Theoretically, this technique is sensitive enough to permit quantification from only one copy of mRNA. Another advantage of using the RT-PCR technique is that it is somewhat tolerant to RNA degradation. This is due to that only a small part of the RNA, which the primers recognize, will be amplified. Furthermore, the cDNA obtained can be stored and used later to screen for every possible transcript of interest. One drawback of using the RT-PCR method is that it requires substantial optimization. Suitable primers have to be designed and since the PCR products increase exponentially with every cycle until a plateau phase is reached, it is important to make sure that the products being analyzed are still in the exponential amplification phase. Another way to overcome this problem is to use real-time PCR technique. In our studies, RT-PCR was chosen to study the differences in cytokine mRNA METHODOLOGICAL CONSIDERATIONS 35 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Figure 8. Schematic illustration of the principle behind RT-PCR. Isolated RNA is reverse transcribed into cDNA. The cDNA is then analyzed for DNA sequences using antisense primers in cycles of amplification. levels since the expression of most cytokine mRNA levels is rather low and hard to detect. Also, as we expect large differences in mRNA expression between treated and non-treated samples, the RT-PCR method is a good choice. The other methods may be preferred if one wants to quantify small differences between different samples. Relative RT-PCR compares transcript abundance of the gene of interest with an internal control. The internal control primers are co-amplified with the gene-specific primers in the same PCR sample. Results are expressed as ratios of the gene specific signal to the internal control signal. This yields a corrected relative value for the gene specific product in each 36 METHODOLOGICAL CONSIDERATIONS L. Fisher sample, accounting for sample differences caused by variable RNA qualities, pipetting or RT efficiency. Thus, this method is considered as a semi-quantitative method. Internal controls commonly used are transcript of constitutively expressed house keeping genes such as β-actin and GAPDH or 18S ribosomal RNA (rRNA). The later have been shown to give less variation in expression due to treatment conditions than the other two; therefore we chose to use 18S rRNA as our internal control. The problem with using rRNA instead of mRNA is that the majority of RNA is rRNA. The endogenous control must be in the same linear range as the mRNA when amplified, or it would otherwise be difficult to detect the PCR product given from the rare mRNA. This problem has been solved by using an identical oligonucleotide sequence as the 18S rRNA primer that has been modified at the 3´end so it cannot be extended by DNA polymerase in the PCR reaction, a system developed by Ambion. These so called competimers are mixed together with the 18S rRNA primers to reduce the amount of PCR product generated from 18S rRNA, leading to the same levels as the cDNA of interest, which can then be detected. 3.6. Analysis of protein expression There are several different ways of detecting specific proteins and changes in protein expression. The most commonly used techniques for protein analysis today are based on immunodetection. The protein acts as an antigen where the antibody recognizes a sequence specific segment or folded conformation of the target protein. The specificity of the antibody-antigen interaction makes it possible to identify a single protein in a mixture of different proteins. The specific protein can be identified by Western blot analysis, immunoprecipitation, immunofluorescence and ELISA (enzyme-linked immunosorbent assay) to mention a few assays. 3.6.1. Western blot In Western blot analysis, protein identification is based on antibody and antigen interactions. In paper IV, we used antibodies directed against APP to analyze APP levels. Western blot analysis is commonly used to positively identify a specific protein in a complex mixture, and to obtain qualitative and quantitative or semi-quantitative data about a protein. It provides details on the proteins apparent molecular weight and processing status. The first step in Western blotting is to separate the individual proteins using gel electrophoresis. The electrophoretic mobility of a protein is determined not only by its molecular weight, but also by its net charge and its shape. For this reason SDS is used, since SDS binds to proteins, disrupts their structure and shape, dissociates them into polypeptide chains, and imposes comparable shapes and net charge densities on the chains. As a result, the proteins are separated only according to their size. Following SDS-polyacryl amide electrophoresis, the separated proteins are transferred or blotted from the gel to a membrane. There are a variety of methods that have been used for this process, including diffusion transfer, capillary transfer, heat-accelerated convectional transfer and vacuum blotting transfer. The transfer method most commonly used for proteins is electrophoretic transfer, because of its speed and transfer efficiency. Here the gel is placed in direct contact with a nitrocellulose membrane or other suitable protein METHODOLOGICAL CONSIDERATIONS 37 Inflammatory Cytokines and NFκB in Alzheimer’s Disease binding support, and placed between two electrodes in a conducting solution. In our experiments, we used a polyvinylidene difluoride membrane. When an electric field is applied, the proteins move out of the gel and onto the membrane in the exact same pattern. Next, the membrane is blocked to reduce the amount of nonspecific binding of antibodies to the surface of the membrane. This is usually done with BSA or non-fat dried milk in solution. After blocking the membrane, it is incubated with labeled primary antibodies or alternatively unlabeled primary antibodies followed by secondary labeled antibodies with subsequent detection. The detection method used is dependent on the label that has been used for the antibody. Labels include biotin, fluorescent probes such as fluorescein or rhodamine, and enzyme conjugates such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). In our studies, we used HRP conjugated secondary antibodies which were detected using enhanced chemiluminescence (ECL). The most common antibody label is enzyme conjugates, which can be detected visually through the conversion of a substrate to a colored precipitate or chemiluminesent light signal at the site of antibody binding. The light output can be captured using a film that is designed for chemiluminescent detection. The intensity of the signal obtained is correlated with the abundance of the protein on the blotting membrane (Fig. 9). Figure 9. Schematic illustration of the Western blot technique with the indirect detection system using ECL. Proteins are separated using gel electrophoresis, transferred to a membrane and detected using labeled antibodies. 3.6.2. ELISA Like with Western blot analysis, ELISA analysis also employs antibody and antigen interactions to identify a specific protein. In paper II, ELISA was used to measure the levels of secreted IL-6 mRNA. ELISA is an extremely sensitive method, and can detect 38 METHODOLOGICAL CONSIDERATIONS L. Fisher an antigen or antibody at a concentration as low as 1 ng/ml. However, the proteins molecular weight cannot be determined by ELISA. The ELISA technique is commonly used to quantify antigens or antibodies using a color change or a flourometric reaction that can be measured quantitatively. In an ELISA, antigens or antibodies first have to be immobilized to a solid surface. Most commonly, this is performed in a 96-well polystyrene plate, which inertly binds antibodies and proteins. The most frequently used ELISA assay is the so called sandwich assay, which is what we used. Here the antigen to be measured is bound between two antibodies, the capture antibody and the detection antibody (Fig. 10). Figure 10. Schematic illustration of the sandwich ELISA technique. The protein is bound between the capture antibody and the enzyme-labeled detection antibody. A colorless substrate will change into a colored product in the presence of the enzyme; wherby the amount of the protein in the sample can be measured. METHODOLOGICAL CONSIDERATIONS 39 Inflammatory Cytokines and NFκB in Alzheimer’s Disease One restriction to this kind of assay is that the antigen has to have more than one antibody binding sites. It is also important that the capture antibody and the detection antibody recognize two non-overlapping epitopes on the antigen. If the antigen is small and only has one epitope, a competitive assay can be performed instead of the sandwich assay. In this case, a labeled antigen and an unlabeled antigen compete for binding to the capture antibody, and a decrease in signal indicates the presence of the antigen in the sample. After binding of the capture antibody to the well surface in the sandwich assay, it is necessary to block the remaining unbound sites of the well with non-specific proteins. Subsequently, the sample mixture is added and the antigen or protein is allowed to bind to the antibody. Next, the labeled detection antibody is bound to the antigen. A detection enzyme may be linked directly to the capture antibody, but with the same disadvantages as for the Western blot analysis, no flexibility in choice of capture antibody label, little signal amplification, and costly. The only advantage of using one antibody would be that it is quicker and the cross-reactivity of the detection antibody is eliminated. Generally, both a capture and a detection antibody are used. The most commonly used enzymes are HRP and AP. In our studies, we used HRP. The enzyme catalyzes a change in a colorless substrate molecule, 1.2-phenylenediaminedihydro-chloride orthophenylene diamine in our case, into a colored product, but the enzyme itself is not consumed in the process, leading to the production of thousands of colored products. This makes the ELISA one of the most sensitive methods for protein detection. The optical density is measured by spectrophotometer to determine the amount of the antibody where the amount of colored product is an indirect expression of the antigen amount in the sample (Fig. 10). If a purified antigen standard is used, the assay can determine the absolute amounts of antigen in an unknown sample. 3.7. Fluorescence microscopy Fluorescence microscopy was used in paper IV for studies on PNA uptake in cultured CGCs and astrocytes. For this purpose the cells were treated with biotinylated PNA, and after permeabilisation, detected by tetramethyl-rhodamine isothiocyanate (TRITC) conjugated streptavidin, which could be visualized under a fluorescence microscope. Biotin (vitamin H) has high affinity for both the glycoprotein avidin and for streptavidin. We preferably used streptavidin because it is less basic and has no carbohydrate residues, resulting in less nonspecific reactions with acidic groups on proteins, as compared to avidin. The strong binding of biotin to fluorochrome conjugated streptavidin then makes indirect visualization of the cells possible. Direct visualization is possible using fluorophores like fluorescein for peptide labeling. However, the streptavidin-biotin approach enables greater flexibility, where different biotinylated molecules can be visualized with the same fluorochrome conjugated streptavidin. Additionally, biotin is a much smaller molecule compared with fluorescein, making it a less disturbing modification of otherwise unmodified PNA. In our study (paper IV), we first fixed the cells with paraformaldehyde before permeabilizing them with Triton X-100. This is to eliminate the possibility that noninternalized PNAs, associated with the plasma membrane, could be translocated into the cell during permeabilization. With alternative fixation methods, like methanol, the cells 40 METHODOLOGICAL CONSIDERATIONS L. Fisher are fixed and permeabilized simultaneously. This enables the PNAs to possibly enter the cell during the procedure, giving false positive results. However, it should be mentioned that recent studies have pointed to the fact that chemical cell fixation may lead to artificial internalization and cytoplasmic localization of peptides bound to the surface of the cell (Leifert et al., 2002; Lundberg and Johansson, 2002; Richard et al., 2003). Therefore, it may also be important to check the cellular distribution of the PNAs in live, unfixed cells, to eliminate possible artifacts. In this case, the PNA should be labeled with a fluorophore. METHODOLOGICAL CONSIDERATIONS 41 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 4. RESULTS AND DISCUSSION 4.1. Cellular delivery of NFκ κB decoy In paper I, we wanted to investigate the possibility to use a cell-penetrating transport peptide to facilitate the translocation of an NFκB decoy across the plasma membrane. In this study, we used the CPPs TP and TP10, both of which have previously been shown to be able to translocate covalently bound larger molecules across the plasma membrane (Pooga et al., 1998a). The transport peptides were linked by a disulfide bond to a cysteine-extended PNA hexamer (PNA(6)) or nonamer (PNA(9)) (for sequences see Table 3). The single-stranded protruding 3´-terminal sequence of the NFκB decoy, complementary to the PNA sequence, was then hybridized to the transport peptide-PNA construct (Fig. 2, paper I). The disulfide bond will, once inside the cell, be reduced leading to dissociation of the PNA-NFκB decoy complex from the transport peptide. This allows the PNA-NFκB decoy, with no interference of the transport peptide, to associate with the NFκB transcription factor. First, we wanted to test the stability of the different transport peptide-PNA-NFκB decoy complexes used. The presence of the various complexes was analyzed by polyacrylamide gel electrophoresis. We found, as expected, that the transport peptide linked to the longer PNA nonamer formed more stable complexes with the NFκB oligonucleotide than the transport peptide linked to the shorter PNA hexamer. The same results could be observed both at room temperature and at 37°C. However, we could not detect any difference in the stability of the complexes formed at room temperature as compared to 37°C using this method. Next, to investigate the effects of the different CPP-PNA-NFκB decoy complexes, we analyzed their abilities to down-regulate the IL-1β-induced NFκB binding activity in Rinm5F cells. Our results showed that, although all complexes used did downregulate the NFκB binding activity, the shorter PNA hexamer together with either one of the transport peptides blocked the NFκB binding activity with lower efficiency. With the use of PNA(9), the NFκB binding activity was almost completely blocked already after 1 h preincubation. In contrast, preincubation with NFκB decoy alone under the same conditions did not result in any significant decrease in NFκB binding activity. Not even an incubation time of 48 h was sufficient to block the NFκB binding activity significantly. In addition, when a downstream effect of blocking NFκB binding activity, namely induction of IL-6 expression, was examined we observed an approx. 20% significant decrease in the IL-1-induced IL-6 mRNA expression. With this, we have shown that the binding of the NFκB decoy to the nonamer PNA linked to TP or TP10, resulted in a stable complex that was efficiently translocated across the plasma membrane. This was the first time, at least to our knowledge, that PNA has been used as a linker in a noncovalent binding between a transport peptide and its cargo. Given that the uptake of DNA into mammalian cells is not very efficient, making high concentrations and long incubation times necessary, this method, using a cell-penetrating transport peptide together with PNA, can be very useful. Using a CPP can in an effective and fast way get an oligonucleotide into a cell. Both the incubation time and the concentration can be lowered to achieve significant effects. Where others have used higher concentrations 42 RESULTS AND DISCUSSION L. Fisher and/or longer incubation times, we have used only 1 µM of the decoy with an incubation time as short as 1 h to obtain significant results. For example, Ye and Johnson (Ye and Johnson, 2001) preincubated with 25 µM of the NFκB decoy for 24 h to attain their results. Furthermore, Tomita and colleagues could after direct transfection with 10 µM NFκB decoy only detect the decoy in about 30% of the cells after 24 h (Tomita et al., 2000c). They could detect the NFκB decoy in about 75% of the cells after using the haemagglutinating virus of Japan (HVJ)–liposome-mediated method of gene transfer, still, 24 h transfection was needed. Further, 12 h of NFκB decoy transfection was necessary to observe any reduction at all of the NFκB binding activity in nuclear extracts from these cells. We are not the only ones using CPPs to transport NFκB inhibitors across the cell membrane. In a study by Takada et al., a CPP was used to deliver a p65 peptide into cells to block NFκB activation (Takada et al., 2004b). However, to suppress the NFκB activation by 25%, 100 µM of the CPP-peptide complex was needed. Further, our approach may also provide a great potential in the capacity to modulate the expression of proteins regulated by other transcription factors. This, because the design of the complexes with an oligonucleotide decoy with a protruding single-stranded 3´-terminal complementary to the PNA sequence, permits the use of the same transport peptide-PNA construct in combination with a decoy containing another consensus sequence. The mechanism involved in the translocation of TP and TP10 into cells, although not fully clarified, has been proposed to be energy- and receptor-independent (Pooga et al., 1998a; Lindgren et al., 2004). Pooga et al. studied the internalization as well as receptor binding for TP in different cell lines. They found that TP localized mainly in the nuclear structure, and that the uptake seemed to be via a non-endocytotic mechanism, since the cellular uptake was temperature independent and unaffected by endocytosis inhibitors. This was in agreement with other studies showing that CPPs enter several different cell lines in a temperature-independent manner (for review see Lindgren et al., 2000). A variety of models have been suggested to describe this energy-independent pathway, including the “pore-formation” model and the “inverted micelle” model (Derossi et al., 1996; Matsuzaki et al., 1996; Matsuzaki et al., 1997; Matsuzaki, 1998). However, since most of the studies done on the mechanism of CPP translocation basically rely on two techniques, fluorescence microscopy on fixed cells or on flow cytometry (FACS) analysis, and that it recently became apparent that the fixation of cells induces artificial cytoplasmic and nuclear localization of CPPs (Pichon et al., 1999; Lundberg and Johansson, 2001; Leifert et al., 2002; Lundberg and Johansson, 2002; Richard et al., 2003), the validity of CPP internalization studies have been questioned. Säälik and colleagues did test this asserted artificial staining upon fixation though, and found no clear redistribution of TP after fixation (Saalik et al., 2004). However, they also noted a reduced uptake of TP at low temperatures and cellular energies, suggesting the involvement of an energy-dependant, endocytotic mechanism. An endocytotic mechanism for cellular internalization has further been proposed for other CPPs (Koppelhus et al., 2002; Console et al., 2003; Drin et al., 2003; Richard et al., 2003; Fischer et al., 2004). So the mechanism, by which CPPs like TP and TP10 translocates into cells, if it is through an energy-independent mechanism or not, remains to be determined. RESULTS AND DISCUSSION 43 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 4.2. IL-1β β - and Aβ β-induced NFκ κB activation Using EMSA, we could in papers II and III detect the activation and nuclear localization of the transcription factor NFκB. The effect of the full length Aβ peptide, Aβ1-42, and the shorter Aβ25-35 on the NFκB binding activity was analyzed. In our study, the Aβ1-42 peptide resulted in a somewhat smaller increase in NFκB binding activity as compared to Aβ25-35 and IL-1β treatments (Fig. 1, paper II). It is possible that the differences observed between the Aβ1-42 peptide and the Aβ25-35 peptide on NFκB binding activity is simply due to the different concentrations used. This is supported by a study from Bonaiuto et al. (Bonaiuto et al., 1997), showing that the same concentration of either Aβ peptides resulted in an equally large induction of NFκB binding activity. However, some studies suggest that the two Aβ peptides may operate via two different mechanisms (Varadarajan et al., 1999; Varadarajan et al., 2001). It cannot be excluded that the amino acid differences between the two peptides could determine their secondary and tertiary conformations, which in turn may bind and activate different glial receptors to induce NFκB activation. As expected we also observed that IL-1β induced NFκB binding activity. Our results are in agreement with other studies showing that both IL-1β and Aβ can stimulate NFκB activation in primary glial cells (Bonaiuto et al., 1997; Akama et al., 1998; Bales et al., 1998; Akama and Van Eldik, 2000). Furthermore, we investigated the subunit composition of the inducible NFκB complexes present in nuclear extracts of activated mixed primary glial cells. Supershift experiments were performed using specific antibodies directed against either the p50 or p65 subunit, confirming the presence of these subunits (Fig.11). This was expected since it has previously been shown that p50/p65 is the main NFκB complex found in glial cells (Bonaiuto et al., 1997; Li et al., 2001). Figure 11. Supershift analysis of nuclear extracts from cells treated with IL-1β (lanes 1-3). The specificity of the bands were shown using antibodies against NFκB subunit p65 (lane 1) and p50 (lane 3). 44 RESULTS AND DISCUSSION L. Fisher In paper III, we showed that co-stimulation of IL-1β together with Aβ25-35 resulted in a significantly higher NFκB binding activity as compared to each of the treatments alone. An additive or synergistic effect was obtained. In AD patients, glial cells that are already primed by Aβ may generate a larger response to inflammatory stimuli than unprimed glial cells in the normal healthy brain. Most AD patients show an ongoing systemic infection in the brain, where systemically secreted cytokines lead to a local higher production of cytokines (Perry et al., 2003). Therefore, the synergistic effect of IL-1β and Aβ co-stimulation observed may possibly reflect the damaging effects that inflammatory processes play in the progression of AD. In another study, we have also shown that both IL-1β and Aβ induce a persistent activation of NFκB, with an early peak at 30 min incubation, which persisted up to 24h (Samuelsson et al., 2005). Since the costimulation of IL-1β and Aβ did not result in a time-dependant decline in NFκB binding activity, we have speculated that the upregulation of activated NFκB observed in AD may be due to that the negative feedback regulation involving induction of the IκB protein by NFκB is not functional in the same way when both Aβ and IL-1β (hence inflammation) is present. Thus, a dysregulation with persistent NFκB activation may be a key contributor to AD. NFκB has for a long time been believed to mediate some of the biological effects induced by Aβ peptides (Bonaiuto et al., 1997; Bales et al., 1998). Aβ has been suggested to interact with RAGE, resulting in activation of NFκB and a triggered proinflammatory pathway (Yan et al., 1996; Du Yan et al., 1997; Schmidt et al., 2000). Therefore, controlling NFκB binding activity may be a useful tool to control the ongoing inflammation that possibly contributes to the progression of AD. 4.3. IL-1β β - and Aβ β-induced upregulation of cytokine expression In view of the data linking Aβ to the induction of several different cytokines, and the fact that the progressive pathology associated with AD has been proposed to at least partly be a consequence of a local inflammatory reaction, we sought to investigate the signaling pathways involved in mediating the induction of cytokines by Aβ peptides. Activated glial cells are believed to contribute, together with Aβ, to the neurotoxicity observed in AD through the production of inflammatory cytokines. Therefore, it was also interesting to investigate the production of inflammatory cytokines by these cells after both Aβ and IL-1β treatment. The IL-1 system has been proposed to play a role in neurodegenerative processes and can, in turn, induce the expression of other cytokines such as IL-6. Also, Aβ peptides have been shown to increase both IL-1β and IL-6 levels (Del Bo et al., 1995; Johnstone et al., 1999; Akama and Van Eldik, 2000; Toro et al., 2001). In paper II and III, we investigated the effects of the Aβ1-42 and Aβ25-35 peptides alone and together with IL-1β on IL-6 and IL-1β expression. Our results indicated that the effect of the Aβ peptides on IL-6 expression was mediated, at least partially, via a pathway not involving induction of IL-1 expression. The mechanism not involving the IL-1-induced expression was indicated by the early induction of IL-6 mRNA levels observed after Aβ treatment, where no IL-1β-induced mRNA levels could yet be detected. Further supporting this was the results on IL-6 secretion, where an additive effect of IL-1β and Aβ25-35 was obtained, and no additional increase in IL-6 secretion RESULTS AND DISCUSSION 45 Inflammatory Cytokines and NFκB in Alzheimer’s Disease could be observed when increasing the IL-1β concentration 10 times. The co-stimulation of Aβ25-35 and IL-1β also resulted in an increased induction of IL-6 mRNA levels, significantly different from IL-1β and Aβ25-35 treatment alone. Moreover, previous studies from our lab have shown that in glial cells from IL-1RI knockout mice, the effect of Aβ25-35 on IL-6 mRNA expression was only partially blocked (Eriksson et al., 1998), additionally supporting that different mechanisms are involved. In addition, a study on APP transgenic mice showed increased levels of IL-6 mRNA expression, but not IL-1β expression (Tehranian et al., 2001), supporting that Aβ can induce IL-6 through a mechanism not involving IL-1β. Our conclusion from these studies is that the induction of IL-6 in the primary glial cell cultures treated with Aβ was not attributed only to increased expression of IL-1β. Further, our results demonstrated that exposure to Aβ25−35 alone resulted in a large increase on IL-6 secretion as compared to non-treated cells. Aβ1-42 also induced a significant, although smaller, increase on IL-6 secretion, as compared to Aβ25−35. The effect of the co-stimulation of Aβ1-42 and IL-1β was not different from the effect induced by IL-1β alone. One might speculate that the IL-6 induction detectable after Aβ treatments is due to that more microglia becomes activated and thereby producing more IL-6 rather than due to a direct effect of Aβ. However, it has previously been shown that IL-6 production is strongly induced by Aβ25-35 in primary astrocytes, but not in primary microglia cells (Forloni et al., 1997). Still, other studies have shown that microglia, as well as monocytic cell lines, also produce IL-6 upon Aβ stimulation (Murphy et al., 1998; Combs et al., 2000; O'Barr and Cooper, 2000) Thus, it cannot be excluded that the microglia cells in the cultures also contributed to the production of IL-6. With regard to the IL-1β treatment of the cells, an upregulation of IL-6 mRNA could be detected already after 2 h, suggesting a direct effect on IL-6 expression, not involving de novo protein synthesis. This was expected since activation of transcription factors induced by IL-1 receptor activation are able to induce transcription of IL-6 (Baeuerle and Henkel, 1994; Lieb et al., 1996). We further observed a marked morphology change of both the astrocytes and microglia cells after incubation with IL-1β and the two different Aβ peptides (Fig. 4, paper II). The network of processes formed by the activated astrocytes with clusters of small and round microglia on top was more pronounced in cultures treated with Aβ25-35 as compared to cultures treated with Aβ1-42. This effect was even more dramatic when the cell cultures were co-stimulated with Aβ and IL-1β. These observations suggest a correlation between morphology and the effects of cytokine secretion. Our results, together with several other studies, suggest that in addition to Aβ peptides other stimuli may also be necessary for maximal effects on IL-6 secretion in glial cells. Co-stimulation of Aβ1-40 or Aβ25-35 together with IL-1β has been shown to strongly increase the secretion of IL-6 in human astrocytoma cell lines (Gitter et al., 1995; Kim et al., 2002). Another study using primary astrocytes showed that the Aβinduced IL-6 production was strongly augmented in the presence of LPS (Forloni et al., 1997). Additionally, a murine microglia cell line was shown to increase the IL-6 production after co-stimulation of Aβ1-40 together with M-CSF (Murphy et al., 1998). Thereof, it can be speculated that accumulation of Aβ peptides in AD brain may activate glial cells to induce IL-6 mRNA. Then, in the presence of an inflammatory stimulus like 46 RESULTS AND DISCUSSION L. Fisher IL-1β, the translation rate of IL-6 mRNA may be increased. As a matter of fact, there are numerous studies indicating that this is the case. Although the mechanism is not known, it has been suggested that activation of two independent intracellular signaling pathways may be necessary for translation of cytokines (Arend et al., 1989; Schindler et al., 1990). As pointed out earlier (section 1.3.5.), increased levels of IL-6 have been detected early in AD brains, and polymorphism in genes encoding IL-6 has been associated with increased risk for AD. Thus, regulating the IL-6 gene in the brain may be of great importance for the control of inflammatory processes observed in AD. Aβ1-40 and LPS co-stimulation has also been demonstrated to synergistically enhance IL-6 expression as well as NO production in a microglia cell line (Gasic-Milenkovic et al., 2003), suggesting that a co-existing stimulus may be necessary for the Aβ-causing damaging effects. Inflammatory cytokines like IL-1β may therefore boost Aβ toxicity or Aβ may augment the proinflammatory signals of IL-1β. Either way, therapies aimed to reduce inflammatory signals may be beneficial in the treatment of AD. 4.4. Inhibition of NFκ κB-binding activity in glial cells In paper I, we showed that by the use of a cell-penetrating transport peptide, cellular uptake of the NFκB decoy was greatly improved. We demonstrated that IL-1β-induced NFκB binding activity could be significantly blocked in rat Rinm5F insulinoma cells, using the transport peptide-PNA(9)-NFκB decoy complex, after only 1 h preincubation at as low concentrations as 1 µM. In paper III, we applied this method to downregulate the NFκB binding activity in mixed primary glial cells. In this study, we decided to use the transport peptide TP10. This was mainly due to studies showing that TP10 does not inhibit basal GTPase activity, as do a lot of CPPs, including TP (Soomets et al., 2000). In addition, the uptake of TP and TP10 into cells have been shown to be equally efficient (Soomets et al., 2000), and our previous results showed that there were no differences in the inhibition of NFκB binding activity using either one of them (paper I). Since, the longer PNA(9) was shown to form a more stable complex with the decoy than PNA(6) in paper I, this PNA was used. We found that the IL-1β- and Aβ-induced NFκB binding activity can be efficiently inhibited using TP10-PNA(9)-NFκB decoy in mixed glial cells. NFκB is one of the key transcription factors regulating the expression of a great variety of genes encoding proteins involved in immune and inflammatory responses (Baldwin, 1996; O'Neill and Kaltschmidt, 1997). The genes encoding the proinflammatory cytokines IL-1β and IL-6 have been shown to be targets of NFκB regulation (Sparacio et al., 1992; Ng et al., 1994; Merola et al., 1996). With the increasing body of evidence, supporting an inflammatory role for NFκB in the brain, blocking this transcription factor may be of great importance. To obstruct NFκB might be a suitable drug target for diseases where an upregulation of inflammatory components is observed. Given the diverse processes involved in activating the NFκB pathway, it is not surprising that a number of different inhibitors can prevent activation of this pathway. Indeed, there have been several lines of strategies used to control NFκB activation and the consequential downstream activating cascade. The first evidence that the NFκB pathway could be specifically inhibited came from studies of IκB mutants (for review see RESULTS AND DISCUSSION 47 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Ghosh et al., 1998). The signal-induced phosphorylation and degradation of IκB were shown to be blocked when overexpressing the IκBα protein with mutations at serine residues 32 and 36, thus preventing the nuclear translocation of NFκB. However, further studies have shown that blocking the NFκB pathway by the IκB repressor, enhances the sensitivity of cells to apoptosis (Van Antwerp et al., 1996; Wang et al., 1996a). Also IKK inhibitors have been shown to interfere with the induced production of NFκB target genes (Roshak et al., 2002; McIntyre et al., 2003). In addition, IKK inhibitors may come with a high risk of toxicity, since mice lacking IKK show embryonic lethality and liver degeneration (Li et al., 1999; Tanaka et al., 1999). Furthermore, anti-inflammatory agents like aspirin and sodium salicylate, (Pierce et al., 1996; Yin et al., 1998; Yamamoto et al., 1999; Takada et al., 2004a), as well as antioxidants (Schreck et al., 1992; Packer and Suzuki, 1993; Nurmi et al., 2004) have been shown to have some restraining effects on NFκB activation. In addition, steroid hormones like glucocorticoids have been reported to have inhibitory effects on NFκB (Auphan et al., 1995; Scheinman et al., 1995; De Bosscher et al., 1997; Sheppard et al., 1998), as do protease and phosphatase inhibitors (Higuchi et al., 1995; Singh and Aggarwal, 1995b; Rossi et al., 1998). However, such compounds have only partial effects on NFκB regulation and can further have undesirable side effects. One approach that is particular attractive in blocking NFκB, which has gained a lot of interest lately, is the use of synthetic ds DNA to act as decoys. This approach has also been shown to be more effective and long-lasting than antisense techniques in blocking constitutively expressed factors like NFκB (Khaled et al., 1998; Dodel et al., 1999). Since NFκB acts as a transcriptional regulator for genes participating in inflammation and cell death, the method we have described may possibly provide for beneficial therapies where inhibiting NFκB may lower these processes in glial cells. Nevertheless, since NFκB activation also has shown to have protective effects in neurons, it may be essential to be able to limit the inhibition of NFκB to only glial cells. To do this, the NFκB decoys used must be designed to distinguish between neuronal and glial NFκB complexes. This should be possible since it has been revealed that the predominant κB-binding factor in neurons was distinct from the bona fide NFκB complex (p65/p50 heterodimer) important in the inflammatory response (Moerman et al., 1999). With the use of CPPs, it might also be possible to direct the CPP-decoy complex to a specific cell type, i.e. glial cells. Thus, designing a specific CPP-NFκB DNA-binding decoy complex that only blocks the effect of NFκB in glial cells, but not in neuronal cells, can be the next big step towards finding new therapies aimed at reducing inflammatory processes triggered by glial cells. 4.5. Decreased IL-6 mRNA expression using NFκ κB decoy Since we previously have shown that IL-1β and Aβ peptides activate NFκB and that this treatment also resulted in an induced IL-6 expression in glial cells, our next step was to investigate the possibility to suppress the IL-6 expression using our NFκB decoy approach. In this study (paper III), we pretreated primary glial cells with the TP10PNA(9)-NFκB decoy complex before stimulation with IL-1β together with Aβ. The 48 RESULTS AND DISCUSSION L. Fisher results demonstrated that preincubation of the cells with TP10-PNA(9)-NFκB decoy significantly decreased the induction of the IL-6 mRNA transcript with approx. 50%. This confirmed that the increased IL-6 production, induced by IL-1β and Aβ, is partly due to increased activity of NFκB. Thus, binding of the NFκB decoy to the NFκB dimer consequently reduced the binding of the NFκB dimer to the promoter region of the IL-6 gene, resulting in a reduction in the IL-6 mRNA expression (Fig. 12). This means that blocking NFκB could possibly be a useful method to suppress the increased levels of IL6 observed in AD. Figure 12. Schematic illustration of the uptake of the transport peptide-PNA-NFκB decoy complex into the cell, with subsequent effect on NFκB target genes. Once inside the cell, the activated NFκB will bind to the decoy, thereby blocking the binding of NFκB to response elements on target genes, and inhibit induction of transcription. Activated NFκB in response to Aβ and inflammatory stimuli may also generate a positive autoregulatory loop where induced cytokines, like IL-1β, also activate NFκB. RESULTS AND DISCUSSION 49 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Thus, it is possible that in AD, NFκB becomes inappropriately activated, causing increased and prolonged expression of proinflammatory genes, leading to pathology. Constant activation of astrocytes and microglia by IL-1β secretion has been shown to inhibit re-outgrowth processes and promote neurodegeneration (Streit et al., 1999). The fact that APP is a target gene for NFκB regulation, and Aβ activates NFκB, may also add to this equation. In fact, increased levels of activated NFκB have been observed in the AD brain (Terai et al., 1996; Boissiere et al., 1997; Kitamura et al., 1997). This may be the cause of the elevated levels of IL-6 expression also observed in AD. It has further been suggested that IL-6 dysregulation and overexpression may contribute to the neuropathology associated with many diseases, including AD (for review see Gadient and Otten, 1997). The increased levels of IL-6 observed in the early stages of AD, prior to Aβ plaque formation, may even be an early primer for the neuropathological changes occurring in AD. It has been demonstrated that exposure of neurons to low levels of IL-6 cause enhanced membrane depolarization, increased intracellular calcium, and enhanced NMDA receptor-mediated neurotoxicity (Qiu et al., 1998). In addition, IL-6 has also been shown to activate a protein kinase proposed to be responsible for the hyperphosphorylation of tau (Rodrigo et al., 2004). Therefore, the increased levels of IL-6 in AD brain may be adequate to prompt individuals to the onset of neurodegeneration. The fact that the IL-6 expression could not be completely blocked by blocking the NFκB binding activity was expected. IL-6 expression is not exclusively regulated through an NFκB-dependent pathway, and the IL-6 promoter contains binding sites for other transcription factors than NFκB. C/EBP, for one, has been shown to be involved in the regulation of IL-6 expression (Akira et al., 1990; Natsuka et al., 1992; Matsusaka et al., 1993; Chandrasekar et al., 1999). Like the NFκB family, the C/EBP family of transcription factors regulates a wide variety of genes involved in inflammation, and they are themselves upregulated by cytokines like IL-1 and IL-6 (Magalini et al., 1995; Cardinaux et al., 2000). In addition, elevated levels of C/EBPδ have been detected in astrocytes in the vicinity of Aβ deposits in AD brains (Li et al., 2004). Since it has been proposed that these two transcription factors (NFκB and C/EBP) work in agreement with each other (see section 1.4), they may both play an important role in the regulation of IL6 in AD. However, it seems like NFκB is central in the regulation of the IL-6 gene and that C/EBP participates in the transcriptional regulation for utmost induction. The AP-1 transcription factor has also been shown to be involved in IL-1β-stimulated IL-6 expression (Hungness et al., 2000). However, NFκB has been considered to be the predominant transcription factor regulating the IL-6 gene in response to inflammatory stimuli (Libermann and Baltimore, 1990; Baeuerle and Henkel, 1994; Vanden Berghe et al., 1999). In conclusion, the results of this study have provided a potential useful tool to reduce the expression of IL-6 and other inflammatory proteins that may be involved in AD. The fact that we could block NFκB and downregulate subsequent IL-6 expression with the use of low concentrations (1µM) and very short incubation times (1 h) is very promising. This should minimize the risk of dangerous and toxic effects that might come with the use of higher doses over prolonged periods of time. It should also be mentioned that in our experiments, we could not detect any remarkably changes in the cell culture morphology in cultures pretreated with TP10-PNA(9)-NFκB decoy as compared to non 50 RESULTS AND DISCUSSION L. Fisher pretreated. If we compare our study with other studies using the decoy approach to block NFκB and followed IL-6 induction (Khaled et al., 1998; Ye and Johnson, 2001), no one has reached the same effectiveness of the decoy with the same exposure time and concentration as we have. The tool that we have developed may be useful not only for regulation of effects of NFκB. Since the design of the transport peptide-PNA oligonucleotide complex permits the use of the same transport peptide-PNA construct in combination with decoys containing sequences for other transcription factors, this may also provide for a great, new therapeutic strategy to reduce or inhibit the expression of proteins involved in other diseases. 4.6. Downregulation of APP in neurons and glial cells using PNA Besides blocking the effects of Aβ, with for instance the use of NFκB decoys, the production of Aβ could also be lowered by downregulating its precursor, APP. In paper IV, we wanted to investigate the possibility to downregulate APP using unmodified antisense PNA, i.e. not coupled to a CPP. Unmodified PNA has been shown to have poor intrinsic cellular uptake properties (Hanvey et al., 1992; Wittung et al., 1995). However, recently, PNA has been reported to have antisense effects in neuronal tissue in vivo (Tyler et al., 1998; Tyler et al., 1999; Rezaei et al., 2001; Tyler-McMahon et al., 2001; McMahon et al., 2002b; Boules et al., 2004). Additionally, studies have indicated that unmodified PNA can enter neuronal cells in culture, probably through an endocytotic mechanism (Aldrian-Herrada et al., 1998). In this study, we used a biotinylated, 15-mer PNA directed to the initiator codon region of rat APP mRNA. Our in vitro studies, comparing neuronal and astrocyte cell cultures, demonstrated that PNA was taken up and could be detected in the cytoplasm of neurons after 4 h incubation. In the astrocytes, on the other hand, no PNA could be detected at this time. After 24 h incubation, PNA could also be observed in the astrocytic cell cultures. The distribution of PNA in the two cell types differed. Even if no nuclear uptake of PNA could be detected in either cell type, the cytoplasmic allocation was more punctuated, vesicle-like in the astrocytes compared to the more diffuse distribution in neurons (Fig. 1 and 2, paper IV). This suggests different mechanisms by which the PNA is taken up by neurons and astrocytes, where a more rapid uptake takes place in neurons. However, more of the astrocytes seem to take up PNA after 24 h as compared to the neurons, approx. 60% and 40%, respectively. When evaluating the effect of PNA treatment on cell survival, no differences were observed in respect to morphological changes or the number of cells as compared with control. We found a concentration-dependent effect of PNA on APP expression in both neurons and astrocytes (Table 4). In both cell types, 3 µM of PNA did not significantly reduce the APP expression, whereas 5 µM did. The most prominent effects were obtained by 10 µM PNA, where we estimated the downregulation of APP expression to approx. 45% and 60% in neurons and glial cells, respectively. This is in accordance with the fact that in astrocytes, the number of cells taken up PNA was higher than in neurons. No effect on APP expression could be observed after treatment with scrambled PNA, demonstrating the specificity of PNA as an antisense agent. Antisense oligonucleotides have previously been used to downregulate APP expression (Majocha et al., 1994; RESULTS AND DISCUSSION 51 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Allinquant et al., 1995; Coulson et al., 1997; Kumar et al., 2000). In vivo studies using 42-mer antisense phosphorothiolated oligonucleotides directed to the Aβ region of APP, showed decreased APP expression (Kumar et al., 2000) as well as Aβ levels (Poon et al., 2004) after intracerebroventricular administration in transgenic mice. A recent study demonstrated that both sense and antisense PNA can downregulate APP in vivo by complementary binding to DNA or mRNA (Boules et al., 2004). They observed about a 70% drop in APP as compared with control. Further, another study using PNA targeted to APP decreased the brain Aβ1-40 and Aβ1-42 in mice with 37% and 47%, respectively (McMahon et al., 2002a). Table 4. APP levels in neurons and astrocytes after PNA treatment. PNA concentration [µm] 0 3 5 10 APP levels in neurons 100.0 99.3 70.7 62.1 APP levels in astrocytes 100.0 83.4 59.2 42.3 APP has been suggested to be involved in neurite outgrowth, synaptic plasticity, and neuroprotection against a wide variety of insults (for review see Panegyres, 2001). In addition, it has been shown that APP knockouts have marked reactive gliosis within brain tissue, and exhibit behavioural deficits suggesting that APP deficient mice may have an impaired neuronal function (Zheng et al., 1995; Dawson et al., 1999; Seabrook et al., 1999). Nevertheless, downregulating APP may still be very useful in the treatment of AD to reduce the amyloid burden. This study showed that unmodified PNA, targeted to the APP mRNA, specifically and effectively downregulated APP in neurons and glial cells in vitro. Our study, together with other studies showing that unmodified PNA can be used in vivo to reduce the levels of APP and Aβ (McMahon et al., 2002a; Boules et al., 2004, paper IV), demonstrates that unmodified PNA can be used as an effective antisense agent in brain cells. 52 RESULTS AND DISCUSSION L. Fisher 5. CONCLUSIONS The question still remains, is the inflammatory reaction a primary cause and driving force of AD pathology, or is it an important participant contributing to AD development, or is the inflammation simply just a consequence and of no importance for the disease progression? Well, multiple lines of evidence collectively point to an early and critical role for inflammation in the development of AD (i.e. glial activation, cytokine overexpression, and effects of gene polymorphism). Maybe the inflammation itself does not underlie the etiology of AD, but it sure looks like it does accompany it. It is possible that it is the combination of inflammatory markers, like cytokines, and a failure in APP metabolism or Aβ clearence, leading to increased levels of Aβ, which are guilty of causing this disease. The potentiation of cytokine secretion by IL-1β together with Aβ (Gitter et al., 1995; Kim et al., 2002; paper II) points in this direction, where the neurodegeneration is mediated, at least in part, by the ability of Aβ to intensify the inflammatory reaction and vice versa. Our studies have clearly shown that Aβ together with IL-1β potentiates the expression of IL-1β and IL-6 as compared with either treatment alone. Our studies have further demonstrated the potentiating effect of Aβ and IL-1β on NFκB activation. Whatever the answer to the question will be, if inflammation is a cause or consequence or something in between, it is still clear that inflammation does indeed occur in the affected brain areas in AD and is associated with Aβ deposition and neuronal damage. Therefore, therapeutic strategies aimed at modulating the inflammatory activity in AD may be a possible cure or at least a possible way to slow down the progression of the disease. There is evidence that NSAIDs work in this regard. However, the use of NSAIDs for the treatment of AD is very controversial. Given the failure of recent clinical trials (Aisen et al., 2003; Reines et al., 2004), the use for NSAIDs in the treatment of AD is still a matter of debate. Besides, the NSAIDs presently used to check inflammation comes with toxic side effects, where the most dangerous one being NSAID-induced gastrointestinal ulceration and bleeding (Langman et al., 1985; Allison et al., 1992; Wilcox et al., 1997; Laine et al., 2003; Clinard et al., 2004). This makes them not very attractive when it comes to prescribing them for chronic treatment, particularly not to the elderly. Steroids, also aimed to control the inflammation in AD patients, have numerous side effects. They have, for example, been shown to be toxic to hippocampal neurons (Sapolsky et al., 1990; Hoschl and Hajek, 2001), which might worsen the pathology in AD. Therefore, it is of great importance to find more specific and selective agents in the treatment of inhibiting the ongoing inflammation that probably contributes to the development of AD. Since NFκB acts as a transcriptional regulator for genes participating in inflammation and cell death, our findings provide new tools to get insights regarding the regulation of NFκB activity for neuronal survival and the mechanisms controlling Aβ production in the brains of AD patients. The present study has provided us with information on a probable drug treatment that can downregulate the proinflammatory cytokines expressed by glial cells, and hopefully reduce inflammatory events occurring in the brain. Since the increase in numbers of activated glial cells expressing proinflammatory cytokines is observed at an early stage of AD, when cognitive decline is still moderate (Arends et al., 2000; Vehmas et al., 2003), targeting glial activation or its consequential secreted products may provide therapeutic options for treatment of AD CONCLUSIONS 53 Inflammatory Cytokines and NFκB in Alzheimer’s Disease patients. Further, in vivo and clinical studies can tell us about the possible use of these transport peptides in introducing decoys into cells to downregulate NFκB and subsequent gene expression. It is of course important to look into the drawbacks that may come with applying this system in clinical studies. Using oligonucleotides as gene therapy strategies have been shown to cause kidney damage and acute hypotension (Srinivasan and Iversen, 1995; Henry et al., 1997). Since these damaging effects were evident at high doses over prolonged periods of time, the use of CPPs, which significantly should decrease the dose of oligonucleotide needed, may show to be the right tool needed for oligonucleotidebased gene therapy. 54 CONCLUSIONS L. Fisher 6. ACKNOWLEDGEMENTS De flesta har sagt det, alla har känt det, det tåls att höras igen - äntligen. Äntligen har jag nått fram till den mest lästa sidan i hela avhandlingen och den sida som känns absolut roligast att skriva – den då jag får tacka alla som på ett eller annat sätt har bidragit till detta arbete! Jag vill börja med att buga och bocka för all inspiration, hjälp, tålamod och dyrbar tid som du har givit mig Kerstin. I allt jobb som du har stått i under åren så har ändå alltid dina doktorander kommit först. Jag har lärt mig otroligt mycket av dig och för det är jag dig evigt tacksam. Ett stort Tack till alla lärare på institutionen för Neurokemi; Ülo, Anders, Anna, Tiit och Mattias. Ni gör våran institutionen till den bästa tänkbara plats att utvecklas för att bli en alla tiders forskare! Stort Tack till Birgitta för att du håller reda på alla oss och för att du alltid är så glad och positiv, och Ulla för alla trevliga pratstunder och uppmuntrande ord. Siv, ett speciellt Tack till dig, inte bara för att du är oumbärlig här på institutionen utan också för allt du lärt mig och för att du är en sån härlig person och bra vän. Alla doktorander på institutionen, de bästa arbetskamrater jag någonsin haft – Tack! Speciellt Tack till alla rumskamrater som kommit och gått under åren, Ove (för mig är du ingen Ränn-Ove), Heléne, Pernilla, Malin och Ulla. Tack för att ni sett till att vi är och förblir mästarna i doktorandernas bowlingtävling. Tack alla i rum 409, Daniel, Ursel, Viktoria, Anna, Pontus, Linda L och Tina, för alla roliga bowlingkvällar och era tappra försök till att slå oss – Sorry guys! Tack till Sandra, Katarina, Peter, Johanna, Helena G, Maria, Anders, Külliki, Jonas, Samir, Katri, Kalle, Mats, Yang, Henrik, Helena M, Mikael och alla ni andra som gjort åren på Neurokemin lite roligare. Mest av allt vill jag Tacka alla i min grupp, för all vänskap, alla skratt och allt roligt vi haft i och utanför labbet. Veronica O – för att du kom som en fräsch fläkt då vi behövde det som mest; Sofia – för att du är en klippa, jobbar som få; Malin – för allt samarbete och för att du alltid ställer upp, denna avhandling skulle vara långt ifrån klar utan dig, och för att du gillar att skvallra, utan dig hade jag inte vetat något om vad som hänt på institutionen det senaste året; Linda A – utan dig har jag dragit hem för länge sen. Tack för att du har stått vid min sida från början till slut, för att du alltid funnits där att prata med – du är guld värd! Jag vill Tacka alla vänner som sett till att jag har haft en fritid och ett liv utanför labbet. Hans och Nicklas – för alla övernattningar och drinky pinky på balkongen. Anna – min egna lilla party pingla, för alla roliga utekvällar och mysiga ridturer. Alla i gamla Sundsvalls kemi gäng, tänk att vi fortfarande håller ihop; Elin – för alla recept och baknings tips och för att du alltid hänger med, Johan – för att du alltid är så go och gla´, Jessica – för allt skoj som vi haft (minns du resan till Turkiet ;o)) och fortfarande har, Mysan – för alla roliga timmar vi spenderat på gymmet, på krogen, på stan ….. och för att jag fick dela lägenhet med dig i början, Katja – för att du alltid finns där och för att du följde med mig som utbytesstudent och därmed förändrade mitt liv. Och Bodil – för att du har varit min vän och alltid hållit reda på mig ända sedan dagis. I would like to Thank my “other family” – The Fishers and my friends “over there”, Mike, Cathy, Ian, Lori, Scott, and all you other guys. Thank you for all the fun and memorable times and for that you every summer made me forget about the work that lies ahead. Min familj, Farmor – Tack för all uppmuntran och alla goda kakor, de har hållit humöret uppe då kvällarna känts långa och dagarna tunga. Mina änglar; Johanna, Viktoria, Lucas och Lisa. Ni har gjort varje resa ”hem” värt det. Anneli och Christer – Tack för att ni alltid ställer upp och för att ni alltid står ut med lillasyster. Lilian och Lennart – Tack för att ni finns och för att ni gör pappa och mamma så lyckliga! Mamma – vad vore jag utan dig? Tack för allt stöd och för att du ÄR den bästa mamma man kan ha! Pappa – du är allt jag någonsin vill bli, Tack för att du är du! Trevor, words are not enough, Thank you for being my best friend and my soul mate – I love you always. Also thanks for proofreading my thesis, even though you thought it was “boring shit”! Slutligen, till mammas hjärta – Olivia – Tack för att du kom till mitt liv och gav det ny mening – du är mitt allt! ACKNOWLEDGEMENTS 55 Inflammatory Cytokines and NFκB in Alzheimer’s Disease 7. REFERENCES AD mutations database. www.molgen.ua.ac.be/ADMutations/ Aisen, P. S.; Schafer, K. A.; Grundman, M.; Pfeiffer, E.; Sano, M.; Davis, K. L.; Farlow, M. R.; Jin, S.; Thomas, R. G.; Thal, L. J. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. Jama 2003, 289, 2819-26. Akama, K. T.; Albanese, C.; Pestell, R. G.; Van Eldik, L. J. Amyloid beta-peptide stimulates nitric oxide production in astrocytes through an NFkappaB-dependent mechanism. Proc Natl Acad Sci U S A 1998, 95, 5795-800. Akama, K. T.; Van Eldik, L. J. Beta-amyloid stimulation of inducible nitric-oxide synthase in astrocytes is interleukin-1beta- and tumor necrosis factor-alpha (TNFalpha)-dependent, and involves a TNFalpha receptor-associated factor- and NFkappaB-inducing kinase-dependent signaling mechanism. J Biol Chem 2000, 275, 7918-24. Akira, S.; Isshiki, H.; Sugita, T.; Tanabe, O.; Kinoshita, S.; Nishio, Y.; Nakajima, T.; Hirano, T.; Kishimoto, T. A nuclear factor for IL-6 expression (NF-IL6) is a member of a C/EBP family. Embo J 1990, 9, 1897-906. Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, G. M.; Cooper, N. R.; Eikelenboom, P.; Emmerling, M.; Fiebich, B. L.; Finch, C. E.; Frautschy, S.; Griffin, W. S.; Hampel, H.; Hull, M.; Landreth, G.; Lue, L.; Mrak, R.; Mackenzie, I. R.; McGeer, P. L.; O'Banion, M. K.; Pachter, J.; Pasinetti, G.; Plata-Salaman, C.; Rogers, J.; Rydel, R.; Shen, Y.; Streit, W.; Strohmeyer, R.; Tooyoma, I.; Van Muiswinkel, F. L.; Veerhuis, R.; Walker, D.; Webster, S.; Wegrzyniak, B.; Wenk, G.; Wyss-Coray, T. Inflammation and Alzheimer's disease. Neurobiol Aging 2000, 21, 383421. Albensi, B. C.; Mattson, M. P. Evidence for the involvement of TNF and NF-kappaB in hippocampal synaptic plasticity. Synapse 2000, 35, 151-9. Aldrian-Herrada, G.; Desarmenien, M. G.; Orcel, H.; Boissin-Agasse, L.; Mery, J.; Brugidou, J.; Rabie, A. A peptide nucleic acid (PNA) is more rapidly internalized in cultured neurons when coupled to a retro-inverso delivery peptide. The antisense activity depresses the target mRNA and protein in magnocellular oxytocin neurons. Nucleic Acids Res 1998, 26, 4910-6. Allinquant, B.; Hantraye, P.; Mailleux, P.; Moya, K.; Bouillot, C.; Prochiantz, A. Downregulation of amyloid precursor protein inhibits neurite outgrowth in vitro. J Cell Biol 1995, 128, 919-27. Allison, M. C.; Howatson, A. G.; Torrance, C. J.; Lee, F. D.; Russell, R. I. Gastrointestinal damage associated with the use of nonsteroidal antiinflammatory drugs. N Engl J Med 1992, 327, 749-54. Aloisi, F.; Care, A.; Borsellino, G.; Gallo, P.; Rosa, S.; Bassani, A.; Cabibbo, A.; Testa, U.; Levi, G.; Peschle, C. Production of hemolymphopoietic cytokines (IL-6, IL-8, colony-stimulating factors) by normal human astrocytes in response to IL-1 beta and tumor necrosis factor-alpha. J Immunol 1992, 149, 2358-66. Altstiel, L. D.; Sperber, K. Cytokines in Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 1991, 15, 481-95. Alzheimer, A. Über eine eigenartige Erkrankung der Hirnrinde. Allgemeine Zeitschrift für Psychiatrie und Psychisch-Gerichtliche Medizin 1907, 64, 146-148 Andriezen, W.L. The neuroglia elements in the human brain. Brain Journal of Medicine 1893, 2, 227-230 Araujo, D. M.; Lapchak, P. A.; Collier, B.; Quirion, R. Localization of interleukin-2 immunoreactivity and interleukin-2 receptors in the rat brain: interaction with the cholinergic system. Brain Res 1989, 498, 257-66. Arend, W. P.; Massoni, R. J.; Niemann, M. A.; Giclas, P. C. Absence of induction of IL-1 production in human monocytes by complement fragments. J Immunol 1989, 142, 173-8. Arends, Y. M.; Duyckaerts, C.; Rozemuller, J. M.; Eikelenboom, P.; Hauw, J. J. Microglia, amyloid and dementia in alzheimer disease. A correlative study. Neurobiol Aging 2000, 21, 39-47. Arenzana-Seisdedos, F.; Thompson, J.; Rodriguez, M. S.; Bachelerie, F.; Thomas, D.; Hay, R. T. Inducible nuclear expression of newly synthesized I kappa B alpha negatively regulates DNA-binding and transcriptional activities of NF-kappa B. Mol Cell Biol 1995, 15, 2689-96. Armogida, M.; Petit, A.; Vincent, B.; Scarzello, S.; da Costa, C. A.; Checler, F. Endogenous beta-amyloid production in presenilin-deficient embryonic mouse fibroblasts. Nat Cell Biol 2001, 3, 1030-3. 56 REFERENCES L. Fisher Arosio, B.; Trabattoni, D.; Galimberti, L.; Bucciarelli, P.; Fasano, F.; Calabresi, C.; Cazzullo, C. L.; Vergani, C.; Annoni, G.; Clerici, M. Interleukin-10 and interleukin-6 gene polymorphisms as risk factors for Alzheimer's disease. Neurobiol Aging 2004, 25, 1009-15. Asai, M.; Hattori, C.; Szabo, B.; Sasagawa, N.; Maruyama, K.; Tanuma, S.; Ishiura, S. Putative function of ADAM9, ADAM10, and ADAM17 as APP alpha-secretase. Biochem Biophys Res Commun 2003, 301, 231-5. Asami-Odaka, A.; Ishibashi, Y.; Kikuchi, T.; Kitada, C.; Suzuki, N. Long amyloid beta-protein secreted from wild-type human neuroblastoma IMR-32 cells. Biochemistry 1995, 34, 10272-8. Aubert, A.; Vega, C.; Dantzer, R.; Goodall, G. Pyrogens specifically disrupt the acquisition of a task involving cognitive processing in the rat. Brain Behav Immun 1995, 9, 129-48. Auphan, N.; DiDonato, J. A.; Rosette, C.; Helmberg, A.; Karin, M. Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science 1995, 270, 286-90. Ayasolla, K.; Khan, M.; Singh, A. K.; Singh, I. Inflammatory mediator and beta-amyloid (25-35)-induced ceramide generation and iNOS expression are inhibited by vitamin E. Free Radic Biol Med 2004, 37, 325-38. Baeuerle, P. A.; Henkel, T. Function and activation of NF-kappa B in the immune system. Annu Rev Immunol 1994, 12, 141-79. Bagli, M.; Papassotiropoulos, A.; Knapp, M.; Jessen, F.; Luise Rao, M.; Maier, W.; Heun, R. Association between an interleukin-6 promoter and 3' flanking region haplotype and reduced Alzheimer's disease risk in a German population. Neurosci Lett 2000, 283, 109-12. Bakalkin, G.; Yakovleva, T.; Terenius, L. NF-kappa B-like factors in the murine brain. Developmentallyregulated and tissue-specific expression. Brain Res Mol Brain Res 1993, 20, 137-46. Baldwin, A. S., Jr. The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol 1996, 14, 649-83. Bales, K. R.; Du, Y.; Dodel, R. C.; Yan, G. M.; Hamilton-Byrd, E.; Paul, S. M. The NF-kappaB/Rel family of proteins mediates Abeta-induced neurotoxicity and glial activation. Brain Res Mol Brain Res 1998, 57, 63-72. Bales, K. R.; Verina, T.; Dodel, R. C.; Du, Y.; Altstiel, L.; Bender, M.; Hyslop, P.; Johnstone, E. M.; Little, S. P.; Cummins, D. J.; Piccardo, P.; Ghetti, B.; Paul, S. M. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat Genet 1997, 17, 263-4. Bamberger, M. E.; Harris, M. E.; McDonald, D. R.; Husemann, J.; Landreth, G. E. A cell surface receptor complex for fibrillar beta-amyloid mediates microglial activation. J Neurosci 2003, 23, 2665-74. Barger, S. W.; Horster, D.; Furukawa, K.; Goodman, Y.; Krieglstein, J.; Mattson, M. P. Tumor necrosis factors alpha and beta protect neurons against amyloid beta-peptide toxicity: evidence for involvement of a kappa B-binding factor and attenuation of peroxide and Ca2+ accumulation. Proc Natl Acad Sci U S A 1995, 92, 9328-32. Barger, S. W.; Mattson, M. P. Induction of neuroprotective kappa B-dependent transcription by secreted forms of the Alzheimer's beta-amyloid precursor. Brain Res Mol Brain Res 1996, 40, 116-26. Barres, B. A. New roles for glia. J Neurosci 1991, 11, 3685-94. Bauer, J.; Strauss, S.; Schreiter-Gasser, U.; Ganter, U.; Schlegel, P.; Witt, I.; Yolk, B.; Berger, M. Interleukin-6 and alpha-2-macroglobulin indicate an acute-phase state in Alzheimer's disease cortices. FEBS Lett 1991, 285, 111-4. Baumann, H.; Gauldie, J. The acute phase response. Immunol Today 1994, 15, 74-80. Beals, C. R.; Clipstone, N. A.; Ho, S. N.; Crabtree, G. R. Nuclear localization of NF-ATc by a calcineurindependent, cyclosporin-sensitive intramolecular interaction. Genes Dev 1997, 11, 824-34. Beg, A. A.; Ruben, S. M.; Scheinman, R. I.; Haskill, S.; Rosen, C. A.; Baldwin, A. S., Jr. I kappa B interacts with the nuclear localization sequences of the subunits of NF-kappa B: a mechanism for cytoplasmic retention. Genes Dev 1992, 6, 1899-913. Begley, R.; Liron, T.; Baryza, J.; Mochly-Rosen, D. Biodistribution of intracellularly acting peptides conjugated reversibly to Tat. Biochem Biophys Res Commun 2004, 318, 949-54. Behl, C. Alzheimer's disease and oxidative stress: implications for novel therapeutic approaches. Prog Neurobiol 1999, 57, 301-23. Behl, C.; Davis, J.; Cole, G. M.; Schubert, D. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun 1992, 186, 944-50. REFERENCES 57 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Behl, C.; Davis, J. B.; Lesley, R.; Schubert, D. Hydrogen peroxide mediates amyloid beta protein toxicity. Cell 1994, 77, 817-27. Behl, C.; Skutella, T.; Lezoualc'h, F.; Post, A.; Widmann, M.; Newton, C. J.; Holsboer, F. Neuroprotection against oxidative stress by estrogens: structure-activity relationship. Mol Pharmacol 1997, 51, 535-41. Benveniste, E. N. Inflammatory cytokines within the central nervous system: sources, function, and mechanism of action. Am J Physiol 1992, 263, C1-16. Benveniste, E. N.; Sparacio, S. M.; Norris, J. G.; Grenett, H. E.; Fuller, G. M. Induction and regulation of interleukin-6 gene expression in rat astrocytes. J Neuroimmunol 1990, 30, 201-12. Benzi, G.; Moretti, A. Are reactive oxygen species involved in Alzheimer's disease? Neurobiol Aging 1995, 16, 661-74. Besedovsky, H. O.; del Rey, A. Immune-neuroendocrine circuits: integrative role of cytokines. Front Neuroendocrinol 1992, 13, 61-94. Betts, J. C.; Cheshire, J. K.; Akira, S.; Kishimoto, T.; Woo, P. The role of NF-kappa B and NF-IL6 transactivating factors in the synergistic activation of human serum amyloid A gene expression by interleukin-1 and interleukin-6. J Biol Chem 1993, 268, 25624-31. Bhakar, A. L.; Tannis, L. L.; Zeindler, C.; Russo, M. P.; Jobin, C.; Park, D. S.; MacPherson, S.; Barker, P. A. Constitutive nuclear factor-kappa B activity is required for central neuron survival. J Neurosci 2002, 22, 8466-75. Bignami, A.; Eng, L. F.; Dahl, D.; Uyeda, C. T. Localization of the glial fibrillary acidic protein in astrocytes by immunofluorescence. Brain Res 1972, 43, 429-35. Bisaglia, M.; Venezia, V.; Biglieri, M.; Russo, C.; Mancini, F.; Milanese, C.; Schettini, G. alphaGlycerylphosphorylethanolamine rescues astrocytes from mitochondrial impairment and oxidative stress induced by amyloid beta-peptides. Neurochem Int 2004, 44, 161-70. Bisaglia, M.; Venezia, V.; Piccioli, P.; Stanzione, S.; Porcile, C.; Russo, C.; Mancini, F.; Milanese, C.; Schettini, G. Acetaminophen protects hippocampal neurons and PC12 cultures from amyloid betapeptides induced oxidative stress and reduces NF-kappaB activation. Neurochem Int 2002, 41, 4354. Bitting, L.; Naidu, A.; Cordell, B.; Murphy, G. M., Jr. Beta-amyloid peptide secretion by a microglial cell line is induced by beta-amyloid-(25-35) and lipopolysaccharide. J Biol Chem 1996, 271, 16084-9. Blalock, J. E. A molecular basis for bidirectional communication between the immune and neuroendocrine systems. Physiol Rev 1989, 69, 1-32. Blanchard, V.; Moussaoui, S.; Czech, C.; Touchet, N.; Bonici, B.; Planche, M.; Canton, T.; Jedidi, I.; Gohin, M.; Wirths, O.; Bayer, T. A.; Langui, D.; Duyckaerts, C.; Tremp, G.; Pradier, L. Time sequence of maturation of dystrophic neurites associated with Abeta deposits in APP/PS1 transgenic mice. Exp Neurol 2003, 184, 247-63. Boissiere, F.; Hunot, S.; Faucheux, B.; Duyckaerts, C.; Hauw, J. J.; Agid, Y.; Hirsch, E. C. Nuclear translocation of NF-kappaB in cholinergic neurons of patients with Alzheimer's disease. Neuroreport 1997, 8, 2849-52. Boland, K.; Behrens, M.; Choi, D.; Manias, K.; Perlmutter, D. H. The serpin-enzyme complex receptor recognizes soluble, nontoxic amyloid-beta peptide but not aggregated, cytotoxic amyloid-beta peptide. J Biol Chem 1996, 271, 18032-44. Boland, K.; Manias, K.; Perlmutter, D. H. Specificity in recognition of amyloid-beta peptide by the serpinenzyme complex receptor in hepatoma cells and neuronal cells. J Biol Chem 1995, 270, 28022-8. Bonaiuto, C.; McDonald, P. P.; Rossi, F.; Cassatella, M. A. Activation of nuclear factor-kappa B by betaamyloid peptides and interferon-gamma in murine microglia. J Neuroimmunol 1997, 77, 51-6. Bonham, M. A.; Brown, S.; Boyd, A. L.; Brown, P. H.; Bruckenstein, D. A.; Hanvey, J. C.; Thomson, S. A.; Pipe, A.; Hassman, F.; Bisi, J. E. An assessment of the antisense properties of RNase Hcompetent and steric-blocking oligomers. Nucleic Acids Res 1995, 23, 1197-203. Bosher, J. M.; Labouesse, M. RNA interference: genetic wand and genetic watchdog. Nat Cell Biol 2000, 2, E31-6. Boules, M.; Williams, K.; Gollatz, E.; Fauq, A.; Richelson, E. Down-regulation of amyloid precursor protein by peptide nucleic acid in vivo. J Mol Neurosci 2004, 24, 123-8. Boutin, H.; LeFeuvre, R. A.; Horai, R.; Asano, M.; Iwakura, Y.; Rothwell, N. J. Role of IL-1alpha and IL1beta in ischemic brain damage. J Neurosci 2001, 21, 5528-34. 58 REFERENCES L. Fisher Braak, H.; Braak, E. Morphological criteria for the recognition of Alzheimer's disease and the distribution pattern of cortical changes related to this disorder. Neurobiol Aging 1994, 15, 355-6; discussion 379-80. Branch, A. D. A good antisense molecule is hard to find. Trends Biochem Sci 1998, 23, 45-50. Breitner, J. C. The role of anti-inflammatory drugs in the prevention and treatment of Alzheimer's disease. Annu Rev Med 1996, 47, 401-11. Breitner, J. C.; Gau, B. A.; Welsh, K. A.; Plassman, B. L.; McDonald, W. M.; Helms, M. J.; Anthony, J. C. Inverse association of anti-inflammatory treatments and Alzheimer's disease: initial results of a cotwin control study. Neurology 1994, 44, 227-32. Breitner, J. C.; Welsh, K. A.; Helms, M. J.; Gaskell, P. C.; Gau, B. A.; Roses, A. D.; Pericak-Vance, M. A.; Saunders, A. M. Delayed onset of Alzheimer's disease with nonsteroidal anti-inflammatory and histamine H2 blocking drugs. Neurobiol Aging 1995, 16, 523-30. Brenneman, D. E.; Schultzberg, M.; Bartfai, T.; Gozes, I. Cytokine regulation of neuronal survival. J Neurochem 1992, 58, 454-60. Breteler, M. M.; Claus, J. J.; van Duijn, C. M.; Launer, L. J.; Hofman, A. Epidemiology of Alzheimer's disease. Epidemiol Rev 1992, 14, 59-82. Bristulf, J.; Gatti, S.; Malinowsky, D.; Bjork, L.; Sundgren, A. K.; Bartfai, T. Interleukin-1 stimulates the expression of type I and type II interleukin-1 receptors in the rat insulinoma cell line Rinm5F; sequencing a rat type II interleukin-1 receptor cDNA. Eur Cytokine Netw 1994, 5, 319-30. Brugg, B.; Dubreuil, Y. L.; Huber, G.; Wollman, E. E.; Delhaye-Bouchaud, N.; Mariani, J. Inflammatory processes induce beta-amyloid precursor protein changes in mouse brain. Proc Natl Acad Sci U S A 1995, 92, 3032-5. Brummelkamp, T. R.; Bernards, R.; Agami, R. A system for stable expression of short interfering RNAs in mammalian cells. Science 2002, 296, 550-3. Burdick, D.; Soreghan, B.; Kwon, M.; Kosmoski, J.; Knauer, M.; Henschen, A.; Yates, J.; Cotman, C.; Glabe, C. Assembly and aggregation properties of synthetic Alzheimer's A4/beta amyloid peptide analogs. J Biol Chem 1992, 267, 546-54. Burns, K.; Martinon, F.; Esslinger, C.; Pahl, H.; Schneider, P.; Bodmer, J. L.; Di Marco, F.; French, L.; Tschopp, J. MyD88, an adapter protein involved in interleukin-1 signaling. J Biol Chem 1998, 273, 12203-9. Butterfield, D. A.; Castegna, A.; Lauderback, C. M.; Drake, J. Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimer's disease brain contribute to neuronal death. Neurobiol Aging 2002, 23, 655-64. Butterfield, D. A.; Hensley, K.; Harris, M.; Mattson, M.; Carney, J. beta-Amyloid peptide free radical fragments initiate synaptosomal lipoperoxidation in a sequence-specific fashion: implications to Alzheimer's disease. Biochem Biophys Res Commun 1994, 200, 710-5. Butterfield, D. A.; Koppal, T.; Subramaniam, R.; Yatin, S. Vitamin E as an antioxidant/free radical scavenger against amyloid beta-peptide-induced oxidative stress in neocortical synaptosomal membranes and hippocampal neurons in culture: insights into Alzheimer's disease. Rev Neurosci 1999, 10, 141-9. Buxbaum, J. D.; Liu, K. N.; Luo, Y.; Slack, J. L.; Stocking, K. L.; Peschon, J. J.; Johnson, R. S.; Castner, B. J.; Cerretti, D. P.; Black, R. A. Evidence that tumor necrosis factor alpha converting enzyme is involved in regulated alpha-secretase cleavage of the Alzheimer amyloid protein precursor. J Biol Chem 1998, 273, 27765-7. Buxbaum, J. D.; Oishi, M.; Chen, H. I.; Pinkas-Kramarski, R.; Jaffe, E. A.; Gandy, S. E.; Greengard, P. Cholinergic agonists and interleukin 1 regulate processing and secretion of the Alzheimer beta/A4 amyloid protein precursor. Proc Natl Acad Sci U S A 1992, 89, 10075-8. Cagnin, A.; Brooks, D. J.; Kennedy, A. M.; Gunn, R. N.; Myers, R.; Turkheimer, F. E.; Jones, T.; Banati, R. B. In-vivo measurement of activated microglia in dementia. Lancet 2001, 358, 461-7. Cai, X. D.; Golde, T. E.; Younkin, S. G. Release of excess amyloid beta protein from a mutant amyloid beta protein precursor. Science 1993, 259, 514-6. Campbell, I. L.; Abraham, C. R.; Masliah, E.; Kemper, P.; Inglis, J. D.; Oldstone, M. B.; Mucke, L. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci U S A 1993, 90, 10061-5. Cardinaux, J. R.; Allaman, I.; Magistretti, P. J. Proinflammatory cytokines induce the transcription factors C/EBPbeta and C/EBPdelta in astrocytes. Glia 2000, 29, 91-7. REFERENCES 59 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Caron, N. J.; Quenneville, S. P.; Tremblay, J. P. Endosome disruption enhances the functional nuclear delivery of Tat-fusion proteins. Biochem Biophys Res Commun 2004, 319, 12-20. Carter, D. A.; Desmarais, E.; Bellis, M.; Campion, D.; Clerget-Darpoux, F.; Brice, A.; Agid, Y.; JaillardSerradt, A.; Mallet, J. More missense in amyloid gene. Nat Genet 1992, 2, 255-6. Casal, C.; Serratosa, J.; Tusell, J. M. Relationship between beta-AP peptide aggregation and microglial activation. Brain Res 2002, 928, 76-84. Chandrasekar, B.; Mitchell, D. H.; Colston, J. T.; Freeman, G. L. Regulation of CCAAT/Enhancer binding protein, interleukin-6, interleukin-6 receptor, and gp130 expression during myocardial ischemia/reperfusion. Circulation 1999, 99, 427-33. Chao, C. C.; Hu, S.; Ehrlich, L.; Peterson, P. K. Interleukin-1 and tumor necrosis factor-alpha synergistically mediate neurotoxicity: involvement of nitric oxide and of N-methyl-D-aspartate receptors. Brain Behav Immun 1995, 9, 355-65. Chao, C. C.; Hu, S.; Frey, W. H., 2nd; Ala, T. A.; Tourtellotte, W. W.; Peterson, P. K. Transforming growth factor beta in Alzheimer's disease. Clin Diagn Lab Immunol 1994, 1, 109-10. Chartier-Harlin, M. C.; Crawford, F.; Houlden, H.; Warren, A.; Hughes, D.; Fidani, L.; Goate, A.; Rossor, M.; Roques, P.; Hardy, J. Early-onset Alzheimer's disease caused by mutations at codon 717 of the beta-amyloid precursor protein gene. Nature 1991, 353, 844-6. Chen, J.; Zhou, Y.; Mueller-Steiner, S.; Chen, L. F.; Kwon, H.; Yi, S.; Mucke, L.; Gan, L. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem 2005, 280, 40364-74. Cheng, B.; Christakos, S.; Mattson, M. P. Tumor necrosis factors protect neurons against metabolicexcitotoxic insults and promote maintenance of calcium homeostasis. Neuron 1994, 12, 139-53. Chiang, C. S.; Stalder, A.; Samimi, A.; Campbell, I. L. Reactive gliosis as a consequence of interleukin-6 expression in the brain: studies in transgenic mice. Dev Neurosci 1994, 16, 212-21. Christie, R. H.; Freeman, M.; Hyman, B. T. Expression of the macrophage scavenger receptor, a multifunctional lipoprotein receptor, in microglia associated with senile plaques in Alzheimer's disease. Am J Pathol 1996, 148, 399-403. Chung, H. M.; Struhl, G. Nicastrin is required for Presenilin-mediated transmembrane cleavage in Drosophila. Nat Cell Biol 2001, 3, 1129-32. Chung, I. Y.; Benveniste, E. N. Tumor necrosis factor-alpha production by astrocytes. Induction by lipopolysaccharide, IFN-gamma, and IL-1 beta. J Immunol 1990, 144, 2999-3007. Citron, M.; Oltersdorf, T.; Haass, C.; McConlogue, L.; Hung, A. Y.; Seubert, P.; Vigo-Pelfrey, C.; Lieberburg, I.; Selkoe, D. J. Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production. Nature 1992, 360, 672-4. Clinard, F.; Sgro, C.; Bardou, M.; Hillon, P.; Dumas, M.; Kreft-Jais, C.; Escousse, A.; Bonithon-Kopp, C. Association between concomitant use of several systemic NSAIDs and an excess risk of adverse drug reaction. A case/non-case study from the French Pharmacovigilance system database. Eur J Clin Pharmacol 2004, 60, 279-83. Colotta, F.; Re, F.; Muzio, M.; Bertini, R.; Polentarutti, N.; Sironi, M.; Giri, J. G.; Dower, S. K.; Sims, J. E.; Mantovani, A. Interleukin-1 type II receptor: a decoy target for IL-1 that is regulated by IL-4. Science 1993, 261, 472-5. Combs, C. K.; Johnson, D. E.; Karlo, J. C.; Cannady, S. B.; Landreth, G. E. Inflammatory mechanisms in Alzheimer's disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J Neurosci 2000, 20, 558-67. Console, S.; Marty, C.; Garcia-Echeverria, C.; Schwendener, R.; Ballmer-Hofer, K. Antennapedia and HIV transactivator of transcription (TAT) "protein transduction domains" promote endocytosis of high molecular weight cargo upon binding to cell surface glycosaminoglycans. J Biol Chem 2003, 278, 35109-14. Coraci, I. S.; Husemann, J.; Berman, J. W.; Hulette, C.; Dufour, J. H.; Campanella, G. K.; Luster, A. D.; Silverstein, S. C.; El-Khoury, J. B. CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer's disease brains and can mediate production of reactive oxygen species in response to beta-amyloid fibrils. Am J Pathol 2002, 160, 101-12. Corbett, J. A.; Wang, J. L.; Sweetland, M. A.; Lancaster, J. R., Jr.; McDaniel, M. L. Interleukin 1 beta induces the formation of nitric oxide by beta-cells purified from rodent islets of Langerhans. Evidence for the beta-cell as a source and site of action of nitric oxide. J Clin Invest 1992, 90, 2384-91. 60 REFERENCES L. Fisher Corder, E. H.; Saunders, A. M.; Strittmatter, W. J.; Schmechel, D. E.; Gaskell, P. C.; Small, G. W.; Roses, A. D.; Haines, J. L.; Pericak-Vance, M. A. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 1993, 261, 921-3. Corey, D. R. Peptide nucleic acids: expanding the scope of nucleic acid recognition. Trends Biotechnol 1997, 15, 224-9. Cottrell, T. R.; Doering, T. L. Silence of the strands: RNA interference in eukaryotic pathogens. Trends Microbiol 2003, 11, 37-43. Coulson, E. J.; Barrett, G. L.; Storey, E.; Bartlett, P. F.; Beyreuther, K.; Masters, C. L. Down-regulation of the amyloid protein precursor of Alzheimer's disease by antisense oligonucleotides reduces neuronal adhesion to specific substrata. Brain Res 1997, 770, 72-80. Cuajungco, M. P.; Goldstein, L. E.; Nunomura, A.; Smith, M. A.; Lim, J. T.; Atwood, C. S.; Huang, X.; Farrag, Y. W.; Perry, G.; Bush, A. I. Evidence that the beta-amyloid plaques of Alzheimer's disease represent the redox-silencing and entombment of abeta by zinc. J Biol Chem 2000, 275, 19439-42. Cui, Y.; Le, Y.; Yazawa, H.; Gong, W.; Wang, J. M. Potential role of the formyl peptide receptor-like 1 (FPRL1) in inflammatory aspects of Alzheimer's disease. J Leukoc Biol 2002, 72, 628-35. Cummings, B. J.; Pike, C. J.; Shankle, R.; Cotman, C. W. Beta-amyloid deposition and other measures of neuropathology predict cognitive status in Alzheimer's disease. Neurobiol Aging 1996, 17, 921-33. Dahlgren, K. N.; Manelli, A. M.; Stine, W. B., Jr.; Baker, L. K.; Krafft, G. A.; LaDu, M. J. Oligomeric and fibrillar species of amyloid-beta peptides differentially affect neuronal viability. J Biol Chem 2002, 277, 32046-53. Davies, S.; Dai, D.; Feldman, I.; Pickett, G.; Leslie, K. K. Identification of a novel mechanism of NFkappaB inactivation by progesterone through progesterone receptors in Hec50co poorly differentiated endometrial cancer cells: induction of A20 and ABIN-2. Gynecol Oncol 2004, 94, 463-70. Davis, J. N., 2nd; Chisholm, J. C. The 'amyloid cascade hypothesis' of AD: decoy or real McCoy? Trends Neurosci 1997, 20, 558-9. Davis, N.; Ghosh, S.; Simmons, D. L.; Tempst, P.; Liou, H. C.; Baltimore, D.; Bose, H. R., Jr. Relassociated pp40: an inhibitor of the rel family of transcription factors. Science 1991, 253, 1268-71. Dawson, G. R.; Seabrook, G. R.; Zheng, H.; Smith, D. W.; Graham, S.; O'Dowd, G.; Bowery, B. J.; Boyce, S.; Trumbauer, M. E.; Chen, H. Y.; Van der Ploeg, L. H.; Sirinathsinghji, D. J. Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the beta-amyloid precursor protein. Neuroscience 1999, 90, 1-13. De Bosscher, K.; Schmitz, M. L.; Vanden Berghe, W.; Plaisance, S.; Fiers, W.; Haegeman, G. Glucocorticoid-mediated repression of nuclear factor-kappaB-dependent transcription involves direct interference with transactivation. Proc Natl Acad Sci U S A 1997, 94, 13504-9. De Strooper, B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-Secretase complex. Neuron 2003, 38, 9-12. De Strooper, B.; Saftig, P.; Craessaerts, K.; Vanderstichele, H.; Guhde, G.; Annaert, W.; Von Figura, K.; Van Leuven, F. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 1998, 391, 387-90. Del Bo, R.; Angeretti, N.; Lucca, E.; De Simoni, M. G.; Forloni, G. Reciprocal control of inflammatory cytokines, IL-1 and IL-6, and beta-amyloid production in cultures. Neurosci Lett 1995, 188, 70-4. Delacourte, A.; Sergeant, N.; Champain, D.; Wattez, A.; Maurage, C. A.; Lebert, F.; Pasquier, F.; David, J. P. Nonoverlapping but synergetic tau and APP pathologies in sporadic Alzheimer's disease. Neurology 2002, 59, 398-407. Demidov, V. V.; Potaman, V. N.; Frank-Kamenetskii, M. D.; Egholm, M.; Buchard, O.; Sonnichsen, S. H.; Nielsen, P. E. Stability of peptide nucleic acids in human serum and cellular extracts. Biochem Pharmacol 1994, 48, 1310-3. Derossi, D.; Calvet, S.; Trembleau, A.; Brunissen, A.; Chassaing, G.; Prochiantz, A. Cell internalization of the third helix of the Antennapedia homeodomain is receptor-independent. J Biol Chem 1996, 271, 18188-93. Derossi, D.; Chassaing, G.; Prochiantz, A. Trojan peptides: the penetratin system for intracellular delivery. Trends Cell Biol 1998, 8, 84-7. Derossi, D.; Joliot, A. H.; Chassaing, G.; Prochiantz, A. The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem 1994, 269, 10444-50. REFERENCES 61 Inflammatory Cytokines and NFκB in Alzheimer’s Disease DeWitt, D. A.; Perry, G.; Cohen, M.; Doller, C.; Silver, J. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer's disease. Exp Neurol 1998, 149, 329-40. Dickson, D. W. The pathogenesis of senile plaques. J Neuropathol Exp Neurol 1997, 56, 321-39. Dickson, D. W.; Farlo, J.; Davies, P.; Crystal, H.; Fuld, P.; Yen, S. H. Alzheimer's disease. A doublelabeling immunohistochemical study of senile plaques. Am J Pathol 1988, 132, 86-101. Dickson, D. W.; Lee, S. C.; Mattiace, L. A.; Yen, S. H.; Brosnan, C. Microglia and cytokines in neurological disease, with special reference to AIDS and Alzheimer's disease. Glia 1993, 7, 75-83. Diehl, J. A.; Hannink, M. Identification of a C/EBP-Rel complex in avian lymphoid cells. Mol Cell Biol 1994, 14, 6635-46. Dinarello, C. A. Biology of interleukin 1. Faseb J 1988, 2, 108-15. Dinarello, C. A. Interleukin-1 and its biologically related cytokines. Adv Immunol 1989, 44, 153-205. Dodel, R. C.; Du, Y.; Bales, K. R.; Gao, F.; Paul, S. M. Sodium salicylate and 17beta-estradiol attenuate nuclear transcription factor NF-kappaB translocation in cultured rat astroglial cultures following exposure to amyloid A beta(1-40) and lipopolysaccharides. J Neurochem 1999, 73, 1453-60. Draczynska-Lusiak, B.; Chen, Y. M.; Sun, A. Y. Oxidized lipoproteins activate NF-kappaB binding activity and apoptosis in PC12 cells. Neuroreport 1998, 9, 527-32. Drin, G.; Cottin, S.; Blanc, E.; Rees, A. R.; Temsamani, J. Studies on the internalization mechanism of cationic cell-penetrating peptides. J Biol Chem 2003, 278, 31192-201. Du Yan, S.; Zhu, H.; Fu, J.; Yan, S. F.; Roher, A.; Tourtellotte, W. W.; Rajavashisth, T.; Chen, X.; Godman, G. C.; Stern, D.; Schmidt, A. M. Amyloid-beta peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc Natl Acad Sci U S A 1997, 94, 5296-301. Duffy, P. E.; Rapport, M.; Graf, L. Glial fibrillary acidic protein and Alzheimer-type senile dementia. Neurology 1980, 30, 778-82. Dunn, S. M.; Coles, L. S.; Lang, R. K.; Gerondakis, S.; Vadas, M. A.; Shannon, M. F. Requirement for nuclear factor (NF)-kappa B p65 and NF-interleukin-6 binding elements in the tumor necrosis factor response region of the granulocyte colony-stimulating factor promoter. Blood 1994, 83, 2469-79. Eanes, E. D.; Glenner, G. G. X-ray diffraction studies on amyloid filaments. J Histochem Cytochem 1968, 16, 673-7. Edbauer, D.; Winkler, E.; Regula, J. T.; Pesold, B.; Steiner, H.; Haass, C. Reconstitution of gammasecretase activity. Nat Cell Biol 2003, 5, 486-8. Egholm, M.; Buchardt, O.; Christensen, L.; Behrens, C.; Freier, S. M.; Driver, D. A.; Berg, R. H.; Kim, S. K.; Norden, B.; Nielsen, P. E. PNA hybridizes to complementary oligonucleotides obeying the Watson-Crick hydrogen-bonding rules. Nature 1993, 365, 566-8. El Khoury, J.; Hickman, S. E.; Thomas, C. A.; Cao, L.; Silverstein, S. C.; Loike, J. D. Scavenger receptormediated adhesion of microglia to beta-amyloid fibrils. Nature 1996, 382, 716-9. Elbashir, S. M.; Lendeckel, W.; Tuschl, T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev 2001, 15, 188-200. Eng, L. F.; Ghirnikar, R. S.; Lee, Y. L. Inflammation in EAE: role of chemokine/cytokine expression by resident and infiltrating cells. Neurochem Res 1996, 21, 511-25. Eriksson, G.; Zetterstrom, M.; Cortes Toro, V.; Bartfai, T.; Iverfeldt, K. Hypersensitive cytokine response to beta-amyloid 25-35 in astroglial cells from IL-1 receptor type I-deficient mice. Int J Mol Med 1998, 1, 201-6. Fattori, E.; Lazzaro, D.; Musiani, P.; Modesti, A.; Alonzi, T.; Ciliberto, G. IL-6 expression in neurons of transgenic mice causes reactive astrocytosis and increase in ramified microglial cells but no neuronal damage. Eur J Neurosci 1995, 7, 2441-9. Fernandez-Madrid, I.; Levy, E.; Marder, K.; Frangione, B. Codon 618 variant of Alzheimer amyloid gene associated with inherited cerebral hemorrhage. Ann Neurol 1991, 30, 730-3. Fernandez-Vizarra, P.; Fernandez, A. P.; Castro-Blanco, S.; Serrano, J.; Bentura, M. L.; Martinez-Murillo, R.; Martinez, A.; Rodrigo, J. Intra- and extracellular Abeta and PHF in clinically evaluated cases of Alzheimer's disease. Histol Histopathol 2004, 19, 823-44. Fischer, B.; Schmoll, H.; Riederer, P.; Bauer, J.; Platt, D.; Popa-Wagner, A. Complement C1q and C3 mRNA expression in the frontal cortex of Alzheimer's patients. J Mol Med 1995, 73, 465-71. Fischer, R.; Kohler, K.; Fotin-Mleczek, M.; Brock, R. A stepwise dissection of the intracellular fate of cationic cell-penetrating peptides. J Biol Chem 2004, 279, 12625-35. 62 REFERENCES L. Fisher Fishman, D.; Faulds, G.; Jeffery, R.; Mohamed-Ali, V.; Yudkin, J. S.; Humphries, S.; Woo, P. The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J Clin Invest 1998, 102, 1369-76. Fitch, M. T.; Doller, C.; Combs, C. K.; Landreth, G. E.; Silver, J. Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma. J Neurosci 1999, 19, 8182-98. Forloni, G.; Demicheli, F.; Giorgi, S.; Bendotti, C.; Angeretti, N. Expression of amyloid precursor protein mRNAs in endothelial, neuronal and glial cells: modulation by interleukin-1. Brain Res Mol Brain Res 1992, 16, 128-34. Forloni, G.; Mangiarotti, F.; Angeretti, N.; Lucca, E.; De Simoni, M. G. Beta-amyloid fragment potentiates IL-6 and TNF-alpha secretion by LPS in astrocytes but not in microglia. Cytokine 1997, 9, 759-62. Francis, R.; McGrath, G.; Zhang, J.; Ruddy, D. A.; Sym, M.; Apfeld, J.; Nicoll, M.; Maxwell, M.; Hai, B.; Ellis, M. C.; Parks, A. L.; Xu, W.; Li, J.; Gurney, M.; Myers, R. L.; Himes, C. S.; Hiebsch, R.; Ruble, C.; Nye, J. S.; Curtis, D. aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 2002, 3, 8597. Fraser, S. P.; Suh, Y. H.; Djamgoz, M. B. Ionic effects of the Alzheimer's disease beta-amyloid precursor protein and its metabolic fragments. Trends Neurosci 1997, 20, 67-72. Frautschy, S. A.; Baird, A.; Cole, G. M. Effects of injected Alzheimer beta-amyloid cores in rat brain. Proc Natl Acad Sci U S A 1991, 88, 8362-6. Frautschy, S. A.; Cole, G. M.; Baird, A. Phagocytosis and deposition of vascular beta-amyloid in rat brains injected with Alzheimer beta-amyloid. Am J Pathol 1992, 140, 1389-99. Frautschy, S. A.; Yang, F.; Irrizarry, M.; Hyman, B.; Saido, T. C.; Hsiao, K.; Cole, G. M. Microglial response to amyloid plaques in APPsw transgenic mice. Am J Pathol 1998, 152, 307-17. Frei, K.; Malipiero, U. V.; Leist, T. P.; Zinkernagel, R. M.; Schwab, M. E.; Fontana, A. On the cellular source and function of interleukin 6 produced in the central nervous system in viral diseases. Eur J Immunol 1989, 19, 689-94. Friedman, W. J. Cytokines regulate expression of the type 1 interleukin-1 receptor in rat hippocampal neurons and glia. Exp Neurol 2001, 168, 23-31. Furukawa, K.; Mattson, M. P. The transcription factor NF-kappaB mediates increases in calcium currents and decreases in NMDA- and AMPA/kainate-induced currents induced by tumor necrosis factoralpha in hippocampal neurons. J Neurochem 1998, 70, 1876-86. Furutani, Y.; Notake, M.; Fukui, T.; Ohue, M.; Nomura, H.; Yamada, M.; Nakamura, S. Complete nucleotide sequence of the gene for human interleukin 1 alpha. Nucleic Acids Res 1986, 14, 316779. Futaki, S.; Suzuki, T.; Ohashi, W.; Yagami, T.; Tanaka, S.; Ueda, K.; Sugiura, Y. Arginine-rich peptides. An abundant source of membrane-permeable peptides having potential as carriers for intracellular protein delivery. J Biol Chem 2001, 276, 5836-40. Gadient, R. A.; Otten, U. H. Interleukin-6 (IL-6)--a molecule with both beneficial and destructive potentials. Prog Neurobiol 1997, 52, 379-90. Games, D.; Adams, D.; Alessandrini, R.; Barbour, R.; Berthelette, P.; Blackwell, C.; Carr, T.; Clemens, J.; Donaldson, T.; Gillespie, F. Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 1995, 373, 523-7. Ganchi, P. A.; Sun, S. C.; Greene, W. C.; Ballard, D. W. I kappa B/MAD-3 masks the nuclear localization signal of NF-kappa B p65 and requires the transactivation domain to inhibit NF-kappa B p65 DNA binding. Mol Biol Cell 1992, 3, 1339-52. Gao, F.; Bales, K. R.; Dodel, R. C.; Liu, J.; Chen, X.; Hample, H.; Farlow, M. R.; Paul, S. M.; Du, Y. NFkappaB mediates IL-1beta-induced synthesis/release of alpha2-macroglobulin in a human glial cell line. Brain Res Mol Brain Res 2002, 105, 108-14. Garcia-Ospina, G. P.; Jimenez-Del Rio, M.; Lopera, F.; Velez-Pardo, C. [Neuronal DNA damage correlates with a positive detection of c-Jun, nuclear factor kB, p53 and Par-4 transcription factors in Alzheimer's disease]. Rev Neurol 2003, 36, 1004-10. Gasic-Milenkovic, J.; Dukic-Stefanovic, S.; Deuther-Conrad, W.; Gartner, U.; Munch, G. Beta-amyloid peptide potentiates inflammatory responses induced by lipopolysaccharide, interferon -gamma and 'advanced glycation endproducts' in a murine microglia cell line. Eur J Neurosci 2003, 17, 813-21. REFERENCES 63 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Gentleman, S. M.; Graham, D. I.; Roberts, G. W. Molecular pathology of head trauma: altered beta APP metabolism and the aetiology of Alzheimer's disease. Prog Brain Res 1993, 96, 237-46. Geula, C.; Wu, C. K.; Saroff, D.; Lorenzo, A.; Yuan, M.; Yankner, B. A. Aging renders the brain vulnerable to amyloid beta-protein neurotoxicity. Nat Med 1998, 4, 827-31. Ghosh, S.; May, M. J.; Kopp, E. B. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998, 16, 225-60. Gibertini, M.; Newton, C.; Friedman, H.; Klein, T. W. Spatial learning impairment in mice infected with Legionella pneumophila or administered exogenous interleukin-1-beta. Brain Behav Immun 1995, 9, 113-28. Giesen, U.; Kleider, W.; Berding, C.; Geiger, A.; Orum, H.; Nielsen, P. E. A formula for thermal stability (Tm) prediction of PNA/DNA duplexes. Nucleic Acids Res 1998, 26, 5004-6. Gil, J.; Esteban, M. Induction of apoptosis by the dsRNA-dependent protein kinase (PKR): mechanism of action. Apoptosis 2000, 5, 107-14. Gitter, B. D.; Cox, L. M.; Rydel, R. E.; May, P. C. Amyloid beta peptide potentiates cytokine secretion by interleukin-1 beta-activated human astrocytoma cells. Proc Natl Acad Sci U S A 1995, 92, 1073841. Giulian, D.; Haverkamp, L. J.; Yu, J. H.; Karshin, W.; Tom, D.; Li, J.; Kirkpatrick, J.; Kuo, L. M.; Roher, A. E. Specific domains of beta-amyloid from Alzheimer plaque elicit neuron killing in human microglia. J Neurosci 1996, 16, 6021-37. Giulian, D.; Lachman, L. B. Interleukin-1 stimulation of astroglial proliferation after brain injury. Science 1985, 228, 497-9. Giulian, D.; Woodward, J.; Young, D. G.; Krebs, J. F.; Lachman, L. B. Interleukin-1 injected into mammalian brain stimulates astrogliosis and neovascularization. J Neurosci 1988a, 8, 2485-90. Giulian, D.; Young, D. G.; Woodward, J.; Brown, D. C.; Lachman, L. B. Interleukin-1 is an astroglial growth factor in the developing brain. J Neurosci 1988b, 8, 709-14. Glenner, G. G.; Wong, C. W. Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem Biophys Res Commun 1984a, 122, 1131-5. Glenner, G. G.; Wong, C. W. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984b, 120, 885-90. Goate, A.; Chartier-Harlin, M. C.; Mullan, M.; Brown, J.; Crawford, F.; Fidani, L.; Giuffra, L.; Haynes, A.; Irving, N.; James, L. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature 1991, 349, 704-6. Goedert, M.; Jakes, R.; Crowther, R. A.; Six, J.; Lubke, U.; Vandermeeren, M.; Cras, P.; Trojanowski, J. Q.; Lee, V. M. The abnormal phosphorylation of tau protein at Ser-202 in Alzheimer disease recapitulates phosphorylation during development. Proc Natl Acad Sci U S A 1993, 90, 5066-70. Goldgaber, D.; Harris, H. W.; Hla, T.; Maciag, T.; Donnelly, R. J.; Jacobsen, J. S.; Vitek, M. P.; Gajdusek, D. C. Interleukin 1 regulates synthesis of amyloid beta-protein precursor mRNA in human endothelial cells. Proc Natl Acad Sci U S A 1989, 86, 7606-10. Goldgaber, D.; Lerman, M. I.; McBride, O. W.; Saffiotti, U.; Gajdusek, D. C. Characterization and chromosomal localization of a cDNA encoding brain amyloid of Alzheimer's disease. Science 1987, 235, 877-80. Goldstein, G. W. The blood-brain barrier: interacters between endothelial cells and astrocytes. Mead Johnson Symp Perinat Dev Med 1987, 15-7. Good, L.; Nielsen, P. E. Antisense inhibition of gene expression in bacteria by PNA targeted to mRNA. Nat Biotechnol 1998, 16, 355-8. Goodman, Y.; Steiner, M. R.; Steiner, S. M.; Mattson, M. P. Nordihydroguaiaretic acid protects hippocampal neurons against amyloid beta-peptide toxicity, and attenuates free radical and calcium accumulation. Brain Res 1994, 654, 171-6. Gordon, M. N.; King, D. L.; Diamond, D. M.; Jantzen, P. T.; Boyett, K. V.; Hope, C. E.; Hatcher, J. M.; DiCarlo, G.; Gottschall, W. P.; Morgan, D.; Arendash, G. W. Correlation between cognitive deficits and Abeta deposits in transgenic APP+PS1 mice. Neurobiol Aging 2001, 22, 377-85. Gotz, J.; Chen, F.; van Dorpe, J.; Nitsch, R. M. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 2001, 293, 1491-5. Gould, E.; Beylin, A.; Tanapat, P.; Reeves, A.; Shors, T. J. Learning enhances adult neurogenesis in the hippocampal formation. Nat Neurosci 1999, 2, 260-5. 64 REFERENCES L. Fisher Gouras, G. K.; Tsai, J.; Naslund, J.; Vincent, B.; Edgar, M.; Checler, F.; Greenfield, J. P.; Haroutunian, V.; Buxbaum, J. D.; Xu, H.; Greengard, P.; Relkin, N. R. Intraneuronal Abeta42 accumulation in human brain. Am J Pathol 2000, 156, 15-20. Gratton, J. P.; Yu, J.; Griffith, J. W.; Babbitt, R. W.; Scotland, R. S.; Hickey, R.; Giordano, F. J.; Sessa, W. C. Cell-permeable peptides improve cellular uptake and therapeutic gene delivery of replicationdeficient viruses in cells and in vivo. Nat Med 2003, 9, 357-62. Gray, E. G.; Paula-Barbosa, M.; Roher, A. Alzheimer's disease: paired helical filaments and cytomembranes. Neuropathol Appl Neurobiol 1987, 13, 91-110. Gray, G. D.; Basu, S.; Wickstrom, E. Transformed and immortalized cellular uptake of oligodeoxynucleoside phosphorothioates, 3'-alkylamino oligodeoxynucleotides, 2'-O-methyl oligoribonucleotides, oligodeoxynucleoside methylphosphonates, and peptide nucleic acids. Biochem Pharmacol 1997, 53, 1465-76. Greenfeder, S. A.; Nunes, P.; Kwee, L.; Labow, M.; Chizzonite, R. A.; Ju, G. Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex. J Biol Chem 1995, 270, 13757-65. Griffin, W. S.; Nicoll, J. A.; Grimaldi, L. M.; Sheng, J. G.; Mrak, R. E. The pervasiveness of interleukin-1 in alzheimer pathogenesis: a role for specific polymorphisms in disease risk. Exp Gerontol 2000, 35, 481-7. Griffin, W. S.; Sheng, J. G.; Roberts, G. W.; Mrak, R. E. Interleukin-1 expression in different plaque types in Alzheimer's disease: significance in plaque evolution. J Neuropathol Exp Neurol 1995, 54, 27681. Griffin, W. S.; Sheng, J. G.; Royston, M. C.; Gentleman, S. M.; McKenzie, J. E.; Graham, D. I.; Roberts, G. W.; Mrak, R. E. Glial-neuronal interactions in Alzheimer's disease: the potential role of a 'cytokine cycle' in disease progression. Brain Pathol 1998, 8, 65-72. Griffin, W. S.; Stanley, L. C.; Ling, C.; White, L.; MacLeod, V.; Perrot, L. J.; White, C. L., 3rd; Araoz, C. Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci U S A 1989, 86, 7611-5. Grilli, M.; Chiu, J. J.; Lenardo, M. J. NF-kappa B and Rel: participants in a multiform transcriptional regulatory system. Int Rev Cytol 1993, 143, 1-62. Grilli, M.; Goffi, F.; Memo, M.; Spano, P. Interleukin-1beta and glutamate activate the NF-kappaB/Rel binding site from the regulatory region of the amyloid precursor protein gene in primary neuronal cultures. J Biol Chem 1996a, 271, 15002-7. Grilli, M.; Pizzi, M.; Memo, M.; Spano, P. Neuroprotection by aspirin and sodium salicylate through blockade of NF-kappaB activation. Science 1996b, 274, 1383-5. Grilli, M.; Ribola, M.; Alberici, A.; Valerio, A.; Memo, M.; Spano, P. Identification and characterization of a kappa B/Rel binding site in the regulatory region of the amyloid precursor protein gene. J Biol Chem 1995, 270, 26774-7. Grimaldi, L. M.; Casadei, V. M.; Ferri, C.; Veglia, F.; Licastro, F.; Annoni, G.; Biunno, I.; De Bellis, G.; Sorbi, S.; Mariani, C.; Canal, N.; Griffin, W. S.; Franceschi, M. Association of early-onset Alzheimer's disease with an interleukin-1alpha gene polymorphism. Ann Neurol 2000, 47, 361-5. Grundke-Iqbal, I.; Iqbal, K.; Quinlan, M.; Tung, Y. C.; Zaidi, M. S.; Wisniewski, H. M. Microtubuleassociated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem 1986, 261, 6084-9. Gruol, D. L.; Nelson, T. E. Physiological and pathological roles of interleukin-6 in the central nervous system. Mol Neurobiol 1997, 15, 307-39. Gu, Y.; Misonou, H.; Sato, T.; Dohmae, N.; Takio, K.; Ihara, Y. Distinct intramembrane cleavage of the βamyloid precursor protein family resembling γ-secretase-like cleavage of Notch. J Biol Chem 2001, 276, 35235-8. Guo, Q.; Robinson, N.; Mattson, M. P. Secreted beta-amyloid precursor protein counteracts the proapoptotic action of mutant presenilin-1 by activation of NF-kappaB and stabilization of calcium homeostasis. J Biol Chem 1998, 273, 12341-51. Gutzmann, H.; Hadler, D. Sustained efficacy and safety of idebenone in the treatment of Alzheimer's disease: update on a 2-year double-blind multicentre study. J Neural Transm Suppl 1998, 54, 30110. REFERENCES 65 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Haass, C.; Hung, A. Y.; Selkoe, D. J. Processing of beta-amyloid precursor protein in microglia and astrocytes favors an internal localization over constitutive secretion. J Neurosci 1991, 11, 378393. Haass, C.; Schlossmacher, M. G.; Hung, A. Y.; Vigo-Pelfrey, C.; Mellon, A.; Ostaszewski, B. L.; Lieberburg, I.; Koo, E. H.; Schenk, D.; Teplow, D. B. Amyloid beta-peptide is produced by cultured cells during normal metabolism. Nature 1992, 359, 322-5. Haga, S.; Akai, K.; Ishii, T. Demonstration of microglial cells in and around senile (neuritic) plaques in the Alzheimer brain. An immunohistochemical study using a novel monoclonal antibody. Acta Neuropathol (Berl) 1989, 77, 569-75. Haga, S.; Ikeda, K.; Sato, M.; Ishii, T. Synthetic Alzheimer amyloid beta/A4 peptides enhance production of complement C3 component by cultured microglial cells. Brain Res 1993, 601, 88-94. Hama, T.; Kushima, Y.; Miyamoto, M.; Kubota, M.; Takei, N.; Hatanaka, H. Interleukin-6 improves the survival of mesencephalic catecholaminergic and septal cholinergic neurons from postnatal, twoweek-old rats in cultures. Neuroscience 1991, 40, 445-52. Hannum, C. H.; Wilcox, C. J.; Arend, W. P.; Joslin, F. G.; Dripps, D. J.; Heimdal, P. L.; Armes, L. G.; Sommer, A.; Eisenberg, S. P.; Thompson, R. C. Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor. Nature 1990, 343, 336-40. Hanvey, J. C.; Peffer, N. J.; Bisi, J. E.; Thomson, S. A.; Cadilla, R.; Josey, J. A.; Ricca, D. J.; Hassman, C. F.; Bonham, M. A.; Au, K. G. Antisense and antigene properties of peptide nucleic acids. Science 1992, 258, 1481-5. Hardy, J. A.; Higgins, G. A. Alzheimer's disease: the amyloid cascade hypothesis. Science 1992, 256, 1845. Harper, J. D.; Lansbury, P. T., Jr. Models of amyloid seeding in Alzheimer's disease and scrapie: mechanistic truths and physiological consequences of the time-dependent solubility of amyloid proteins. Annu Rev Biochem 1997, 66, 385-407. Harper, J. D.; Wong, S. S.; Lieber, C. M.; Lansbury, P. T. Observation of metastable Abeta amyloid protofibrils by atomic force microscopy. Chem Biol 1997, 4, 119-25. Harris, M. E.; Carney, J. M.; Cole, P. S.; Hensley, K.; Howard, B. J.; Martin, L.; Bummer, P.; Wang, Y.; Pedigo, N. W., Jr.; Butterfield, D. A. beta-Amyloid peptide-derived, oxygen-dependent free radicals inhibit glutamate uptake in cultured astrocytes: implications for Alzheimer's disease. Neuroreport 1995a, 6, 1875-9. Harris, M. E.; Hensley, K.; Butterfield, D. A.; Leedle, R. A.; Carney, J. M. Direct evidence of oxidative injury produced by the Alzheimer's beta-amyloid peptide (1-40) in cultured hippocampal neurons. Exp Neurol 1995b, 131, 193-202. Hartley, D. M.; Walsh, D. M.; Ye, C. P.; Diehl, T.; Vasquez, S.; Vassilev, P. M.; Teplow, D. B.; Selkoe, D. J. Protofibrillar intermediates of amyloid beta-protein induce acute electrophysiological changes and progressive neurotoxicity in cortical neurons. J Neurosci 1999, 19, 8876-84. Haskill, S.; Beg, A. A.; Tompkins, S. M.; Morris, J. S.; Yurochko, A. D.; Sampson-Johannes, A.; Mondal, K.; Ralph, P.; Baldwin, A. S., Jr. Characterization of an immediate-early gene induced in adherent monocytes that encodes I kappa B-like activity. Cell 1991, 65, 1281-9. Hatten, M. E. Neuronal regulation of astroglial morphology and proliferation in vitro. J Cell Biol 1985, 100, 384-96. Hatten, M. E. Riding the glial monorail: a common mechanism for glial-guided neuronal migration in different regions of the developing mammalian brain. Trends Neurosci 1990, 13, 179-84. Hedley, R.; Hallmayer, J.; Groth, D. M.; Brooks, W. S.; Gandy, S. E.; Martins, R. N. Association of interleukin-1 polymorphisms with Alzheimer's disease in Australia. Ann Neurol 2002, 51, 795-7. Hendriks, L.; van Duijn, C. M.; Cras, P.; Cruts, M.; Van Hul, W.; van Harskamp, F.; Warren, A.; McInnis, M. G.; Antonarakis, S. E.; Martin, J. J. Presenile dementia and cerebral haemorrhage linked to a mutation at codon 692 of the beta-amyloid precursor protein gene. Nat Genet 1992, 1, 218-21. Henkel, T.; Zabel, U.; van Zee, K.; Muller, J. M.; Fanning, E.; Baeuerle, P. A. Intramolecular masking of the nuclear location signal and dimerization domain in the precursor for the p50 NF-kappa B subunit. Cell 1992, 68, 1121-33. Henry, S. P.; Bolte, H.; Auletta, C.; Kornbrust, D. J. Evaluation of the toxicity of ISIS 2302, a phosphorothioate oligonucleotide, in a four-week study in cynomolgus monkeys. Toxicology 1997, 120, 145-55. 66 REFERENCES L. Fisher Hensley, K.; Carney, J. M.; Mattson, M. P.; Aksenova, M.; Harris, M.; Wu, J. F.; Floyd, R. A.; Butterfield, D. A. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease. Proc Natl Acad Sci U S A 1994, 91, 3270-4. Hensley, K.; Hall, N.; Subramaniam, R.; Cole, P.; Harris, M.; Aksenov, M.; Aksenova, M.; Gabbita, S. P.; Wu, J. F.; Carney, J. M.; et al. Brain regional correspondence between Alzheimer's disease histopathology and biomarkers of protein oxidation. J Neurochem 1995, 65, 2146-56. Heutink, P. Untangling tau-related dementia. Hum Mol Genet 2000, 9, 979-86. Heyser, C. J.; Masliah, E.; Samimi, A.; Campbell, I. L.; Gold, L. H. Progressive decline in avoidance learning paralleled by inflammatory neurodegeneration in transgenic mice expressing interleukin 6 in the brain. Proc Natl Acad Sci U S A 1997, 94, 1500-5. Hickey, W. F.; Vass, K.; Lassmann, H. Bone marrow-derived elements in the central nervous system: an immunohistochemical and ultrastructural survey of rat chimeras. J Neuropathol Exp Neurol 1992, 51, 246-56. Higuchi, M.; Singh, S.; Chan, H.; Aggarwal, B. B. Protease inhibitors differentially regulate tumor necrosis factor-induced apoptosis, nuclear factor-kappa B activation, cytotoxicity, and differentiation. Blood 1995, 86, 2248-56. Hilbich, C.; Kisters-Woike, B.; Reed, J.; Masters, C. L.; Beyreuther, K. Aggregation and secondary structure of synthetic amyloid beta A4 peptides of Alzheimer's disease. J Mol Biol 1991, 218, 14963. Hof, P. R.; Giannakopoulos, P.; Vickers, J. C.; Bouras, C.; Morrison, J. H. The morphologic and neurochemical basis of dementia: aging, hierarchical patterns of lesion distribution and vulnerable neuronal phenotype. Rev Neurosci 1995, 6, 97-124. Holcomb, L.; Gordon, M. N.; McGowan, E.; Yu, X.; Benkovic, S.; Jantzen, P.; Wright, K.; Saad, I.; Mueller, R.; Morgan, D.; Sanders, S.; Zehr, C.; O'Campo, K.; Hardy, J.; Prada, C. M.; Eckman, C.; Younkin, S.; Hsiao, K.; Duff, K. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat Med 1998, 4, 97100. Holliday, J.; Gruol, D. L. Cytokine stimulation increases intracellular calcium and alters the response to quisqualate in cultured cortical astrocytes. Brain Res 1993, 621, 233-41. Holtzman, D. M.; Bales, K. R.; Tenkova, T.; Fagan, A. M.; Parsadanian, M.; Sartorius, L. J.; Mackey, B.; Olney, J.; McKeel, D.; Wozniak, D.; Paul, S. M. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2000, 97, 2892-7. Hoozemans, J. J.; Veerhuis, R.; Rozemuller, A. J.; Eikelenboom, P. Non-steroidal anti-inflammatory drugs and cyclooxygenase in Alzheimer's disease. Curr Drug Targets 2003, 4, 461-8. Hoschl, C.; Hajek, T. Hippocampal damage mediated by corticosteroids--a neuropsychiatric research challenge. Eur Arch Psychiatry Clin Neurosci 2001, 251 Suppl 2, II81-8. Howl, J.; Jones, S.; Farquhar, M. Intracellular delivery of bioactive peptides to RBL-2H3 cells induces beta-hexosaminidase secretion and phospholipase D activation. Chembiochem 2003, 4, 1312-6. Hsiao, K.; Chapman, P.; Nilsen, S.; Eckman, C.; Harigaya, Y.; Younkin, S.; Yang, F.; Cole, G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996, 274, 99102. Huang, X.; Cuajungco, M. P.; Atwood, C. S.; Hartshorn, M. A.; Tyndall, J. D.; Hanson, G. R.; Stokes, K. C.; Leopold, M.; Multhaup, G.; Goldstein, L. E.; Scarpa, R. C.; Saunders, A. J.; Lim, J.; Moir, R. D.; Glabe, C.; Bowden, E. F.; Masters, C. L.; Fairlie, D. P.; Tanzi, R. E.; Bush, A. I. Cu(II) potentiation of alzheimer abeta neurotoxicity. Correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem 1999, 274, 37111-6. Huell, M.; Strauss, S.; Volk, B.; Berger, M.; Bauer, J. Interleukin-6 is present in early stages of plaque formation and is restricted to the brains of Alzheimer's disease patients. Acta Neuropathol (Berl) 1995, 89, 544-51. Hull, M.; Fiebich, B. L.; Lieb, K.; Strauss, S.; Berger, S. S.; Volk, B.; Bauer, J. Interleukin-6-associated inflammatory processes in Alzheimer's disease: new therapeutic options. Neurobiol Aging 1996a, 17, 795-800. Hull, M.; Strauss, S.; Berger, M.; Volk, B.; Bauer, J. The participation of interleukin-6, a stress-inducible cytokine, in the pathogenesis of Alzheimer's disease. Behav Brain Res 1996b, 78, 37-41. REFERENCES 67 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Hungness, E. S.; Pritts, T. A.; Luo, G. J.; Sun, X.; Penner, C. G.; Hasselgren, P. O. The transcription factor activator protein-1 is activated and interleukin-6 production is increased in interleukin-1betastimulated human enterocytes. Shock 2000, 14, 386-91. Hussain, I.; Powell, D.; Howlett, D. R.; Tew, D. G.; Meek, T. D.; Chapman, C.; Gloger, I. S.; Murphy, K. E.; Southan, C. D.; Ryan, D. M.; Smith, T. S.; Simmons, D. L.; Walsh, F. S.; Dingwall, C.; Christie, G. Identification of a novel aspartic protease (Asp 2) as beta-secretase. Mol Cell Neurosci 1999, 14, 419-27. Hutchison, D. L.; Myers, R. L. Prostaglandin-mediated suppression of macrophage phagocytosis of Listeria monocytogenes. Cell Immunol 1987, 110, 68-76. Ignatovich, I. A.; Dizhe, E. B.; Pavlotskaya, A. V.; Akifiev, B. N.; Burov, S. V.; Orlov, S. V.; Perevozchikov, A. P. Complexes of plasmid DNA with basic domain 47-57 of the HIV-1 Tat protein are transferred to mammalian cells by endocytosis-mediated pathways. J Biol Chem 2003, 278, 42625-36. Ii, M.; Sunamoto, M.; Ohnishi, K.; Ichimori, Y. beta-Amyloid protein-dependent nitric oxide production from microglial cells and neurotoxicity. Brain Res 1996, 720, 93-100. in t' Veld, B. A.; Ruitenberg, A.; Hofman, A.; Launer, L. J.; van Duijn, C. M.; Stijnen, T.; Breteler, M. M.; Stricker, B. H. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer's disease. N Engl J Med 2001, 345, 1515-21. Irizarry, M. C.; McNamara, M.; Fedorchak, K.; Hsiao, K.; Hyman, B. T. APPSw transgenic mice develop age-related A beta deposits and neuropil abnormalities, but no neuronal loss in CA1. J Neuropathol Exp Neurol 1997, 56, 965-73. Iversen, L. L.; Mortishire-Smith, R. J.; Pollack, S. J.; Shearman, M. S. The toxicity in vitro of beta-amyloid protein. Biochem J 1995, 311 ( Pt 1), 1-16. Iwasaki, Y.; Asai, M.; Yoshida, M.; Nigawara, T.; Kambayashi, M.; Nakashima, N. Dehydroepiandrosterone-sulfate inhibits nuclear factor-kappaB-dependent transcription in hepatocytes, possibly through antioxidant effect. J Clin Endocrinol Metab 2004, 89, 3449-54. Janzer, R. C.; Raff, M. C. Astrocytes induce blood-brain barrier properties in endothelial cells. Nature 1987, 325, 253-7. Jarrett, J. T.; Berger, E. P.; Lansbury, P. T., Jr. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease. Biochemistry 1993, 32, 4693-7. Jensen, K. K.; Orum, H.; Nielsen, P. E.; Norden, B. Kinetics for hybridization of peptide nucleic acids (PNA) with DNA and RNA studied with the BIAcore technique. Biochemistry 1997, 36, 5072-7. Johnson, C.; Van Antwerp, D.; Hope, T. J. An N-terminal nuclear export signal is required for the nucleocytoplasmic shuttling of IkappaBalpha. Embo J 1999, 18, 6682-93. Johnstone, M.; Gearing, A. J.; Miller, K. M. A central role for astrocytes in the inflammatory response to beta-amyloid; chemokines, cytokines and reactive oxygen species are produced. J Neuroimmunol 1999, 93, 182-93. Jones, C. T.; Morris, S.; Yates, C. M.; Moffoot, A.; Sharpe, C.; Brock, D. J.; St Clair, D. Mutation in codon 713 of the beta amyloid precursor protein gene presenting with schizophrenia. Nat Genet 1992, 1, 306-9. Jordan, F. L.; Thomas, W. E. Brain macrophages: questions of origin and interrelationship. Brain Res 1988, 472, 165-78. Kalback, W.; Watson, M. D.; Kokjohn, T. A.; Kuo, Y. M.; Weiss, N.; Luehrs, D. C.; Lopez, J.; Brune, D.; Sisodia, S. S.; Staufenbiel, M.; Emmerling, M.; Roher, A. E. APP transgenic mice Tg2576 accumulate Abeta peptides that are distinct from the chemically modified and insoluble peptides deposited in Alzheimer's disease senile plaques. Biochemistry 2002, 41, 922-8. Kalman, J.; Juhasz, A.; Laird, G.; Dickens, P.; Jardanhazy, T.; Rimanoczy, A.; Boncz, I.; Parry-Jones, W. L.; Janka, Z. Serum interleukin-6 levels correlate with the severity of dementia in Down syndrome and in Alzheimer's disease. Acta Neurol Scand 1997, 96, 236-40. Kaltschmidt, B.; Heinrich, M.; Kaltschmidt, C. Stimulus-dependent activation of NF-kappaB specifies apoptosis or neuroprotection in cerebellar granule cells. Neuromolecular Med 2002, 2, 299-309. Kaltschmidt, B.; Kaltschmidt, C.; Hofmann, T. G.; Hehner, S. P.; Droge, W.; Schmitz, M. L. The pro- or anti-apoptotic function of NF-kappaB is determined by the nature of the apoptotic stimulus. Eur J Biochem 2000, 267, 3828-35. 68 REFERENCES L. Fisher Kaltschmidt, B.; Uherek, M.; Volk, B.; Baeuerle, P. A.; Kaltschmidt, C. Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proc Natl Acad Sci U S A 1997, 94, 2642-7. Kaltschmidt, C.; Kaltschmidt, B.; Baeuerle, P. A. Brain synapses contain inducible forms of the transcription factor NF-kappa B. Mech Dev 1993, 43, 135-47. Kaltschmidt, C.; Kaltschmidt, B.; Lannes-Vieira, J.; Kreutzberg, G. W.; Wekerle, H.; Baeuerle, P. A.; Gehrmann, J. Transcription factor NF-kappa B is activated in microglia during experimental autoimmune encephalomyelitis. J Neuroimmunol 1994a, 55, 99-106. Kaltschmidt, C.; Kaltschmidt, B.; Neumann, H.; Wekerle, H.; Baeuerle, P. A. Constitutive NF-kappa B activity in neurons. Mol Cell Biol 1994b, 14, 3981-92. Kamenetz, F.; Tomita, T.; Hsieh, H.; Seabrook, G.; Borchelt, D.; Iwatsubo, T.; Sisodia, S.; Malinow, R. APP processing and synaptic function. Neuron 2003, 37, 925-37. Kamino, K.; Orr, H. T.; Payami, H.; Wijsman, E. M.; Alonso, M. E.; Pulst, S. M.; Anderson, L.; O'Dahl, S.; Nemens, E.; White, J. A. Linkage and mutational analysis of familial Alzheimer disease kindreds for the APP gene region. Am J Hum Genet 1992, 51, 998-1014. Kang, J.; Lemaire, H. G.; Unterbeck, A.; Salbaum, J. M.; Masters, C. L.; Grzeschik, K. H.; Multhaup, G.; Beyreuther, K.; Muller-Hill, B. The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor. Nature 1987, 325, 733-6. Karin, M. How NF-kappaB is activated: the role of the IkappaB kinase (IKK) complex. Oncogene 1999, 18, 6867-74. Katzman, R.; Saitoh, T. Advances in Alzheimer's disease. Faseb J 1991, 5, 278-86. Kawamura, I.; Morishita, R.; Tomita, N.; Lacey, E.; Aketa, M.; Tsujimoto, S.; Manda, T.; Tomoi, M.; Kida, I.; Higaki, J.; Kaneda, Y.; Shimomura, K.; Ogihara, T. Intratumoral injection of oligonucleotides to the NF kappa B binding site inhibits cachexia in a mouse tumor model. Gene Ther 1999, 6, 91-7. Kawamura, I.; Morishita, R.; Tsujimoto, S.; Manda, T.; Tomoi, M.; Tomita, N.; Goto, T.; Ogihara, T.; Kaneda, Y. Intravenous injection of oligodeoxynucleotides to the NF-kappaB binding site inhibits hepatic metastasis of M5076 reticulosarcoma in mice. Gene Ther 2001, 8, 905-12. Khaled, A. R.; Butfiloski, E. J.; Sobel, E. S.; Schiffenbauer, J. Use of phosphorothioate-modified oligodeoxynucleotides to inhibit NF-kappaB expression and lymphocyte function. Clin Immunol Immunopathol 1998, 86, 170-9. Kilk, K.; Elmquist, A.; Saar, K.; Pooga, M.; Land, T.; Bartfai, T.; Soomets, U.; Langel, Ü. Targeting of antisense PNA oligomers to human galanin receptor type 1 mRNA. Neuropeptides 2004, 38, 31624. Kim, J. Y.; Kim, H.; Lee, S. G.; Choi, B. H.; Kim, Y. H.; Huh, P. W.; Lee, K. H.; Han, H.; Rha, H. K. Amyloid beta peptide (Abeta42) activates PLC-delta1 promoter through the NF-kappaB binding site. Biochem Biophys Res Commun 2003, 310, 904-9. Kim, M. S.; Sung, M. J.; Seo, S. B.; Yoo, S. J.; Lim, W. K.; Kim, H. M. Water-soluble chitosan inhibits the production of proinflammatory cytokine in human astrocytoma cells activated by amyloid beta peptide and interleukin-1beta. Neurosci Lett 2002, 321, 105-9. Kinugawa, K.; Shimizu, T.; Yao, A.; Kohmoto, O.; Serizawa, T.; Takahashi, T. Transcriptional regulation of inducible nitric oxide synthase in cultured neonatal rat cardiac myocytes. Circ Res 1997, 81, 911-21. Kitaguchi, N.; Takahashi, Y.; Tokushima, Y.; Shiojiri, S.; Ito, H. Novel precursor of Alzheimer's disease amyloid protein shows protease inhibitory activity. Nature 1988, 331, 530-2. Kitamura, Y.; Shimohama, S.; Ota, T.; Matsuoka, Y.; Nomura, Y.; Taniguchi, T. Alteration of transcription factors NF-kappaB and STAT1 in Alzheimer's disease brains. Neurosci Lett 1997, 237, 17-20. Kitazawa, M.; Oddo, S.; Yamasaki, T. R.; Green, K. N.; LaFerla, F. M. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer's disease. J Neurosci 2005, 25, 8843-53. Klegeris, A.; McGeer, P. L. beta-amyloid protein enhances macrophage production of oxygen free radicals and glutamate. J Neurosci Res 1997, 49, 229-35. Klegeris, A.; Walker, D. G.; McGeer, P. L. Activation of macrophages by Alzheimer beta amyloid peptide. Biochem Biophys Res Commun 1994, 199, 984-91. Klein, W. L.; Krafft, G. A.; Finch, C. E. Targeting small Abeta oligomers: the solution to an Alzheimer's disease conundrum? Trends Neurosci 2001, 24, 219-24. REFERENCES 69 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Knudsen, H.; Nielsen, P. E. Antisense properties of duplex- and triplex-forming PNAs. Nucleic Acids Res 1996, 24, 494-500. Koh, J. Y.; Yang, L. L.; Cotman, C. W. Beta-amyloid protein increases the vulnerability of cultured cortical neurons to excitotoxic damage. Brain Res 1990, 533, 315-20. Koike, H.; Tomioka, S.; Sorimachi, H.; Saido, T. C.; Maruyama, K.; Okuyama, A.; Fujisawa-Sehara, A.; Ohno, S.; Suzuki, K.; Ishiura, S. Membrane-anchored metalloprotease MDC9 has an alphasecretase activity responsible for processing the amyloid precursor protein. Biochem J 1999, 343 Pt 2, 371-5. Kopec, K. K.; Carroll, R. T. Alzheimer's beta-amyloid peptide 1-42 induces a phagocytic response in murine microglia. J Neurochem 1998, 71, 2123-31. Kopp, E. B.; Ghosh, S. NF-kappa B and rel proteins in innate immunity. Adv Immunol 1995, 58, 1-27. Koppal, T.; Drake, J.; Yatin, S.; Jordan, B.; Varadarajan, S.; Bettenhausen, L.; Butterfield, D. A. Peroxynitrite-induced alterations in synaptosomal membrane proteins: insight into oxidative stress in Alzheimer's disease. J Neurochem 1999, 72, 310-7. Koppelhus, U.; Awasthi, S. K.; Zachar, V.; Holst, H. U.; Ebbesen, P.; Nielsen, P. E. Cell-dependent differential cellular uptake of PNA, peptides, and PNA-peptide conjugates. Antisense Nucleic Acid Drug Dev 2002, 12, 51-63. Koppelhus, U.; Nielsen, P. E. Cellular delivery of peptide nucleic acid (PNA). Adv Drug Deliv Rev 2003, 55, 267-80. Kowall, N. W.; McKee, A. C.; Yankner, B. A.; Beal, M. F. In vivo neurotoxicity of beta-amyloid [beta(140)] and the beta(25-35) fragment. Neurobiol Aging 1992, 13, 537-42. Kreutzberg, G. W. Microglia: a sensor for pathological events in the CNS. Trends Neurosci 1996, 19, 3128. Kumar, V. B.; Farr, S. A.; Flood, J. F.; Kamlesh, V.; Franko, M.; Banks, W. A.; Morley, J. E. Site-directed antisense oligonucleotide decreases the expression of amyloid precursor protein and reverses deficits in learning and memory in aged SAMP8 mice. Peptides 2000, 21, 1769-75. Kunsch, C.; Ruben, S. M.; Rosen, C. A. Selection of optimal kappa B/Rel DNA-binding motifs: interaction of both subunits of NF-kappa B with DNA is required for transcriptional activation. Mol Cell Biol 1992, 12, 4412-21. Laine, L.; Connors, L. G.; Reicin, A.; Hawkey, C. J.; Burgos-Vargas, R.; Schnitzer, T. J.; Yu, Q.; Bombardier, C. Serious lower gastrointestinal clinical events with nonselective NSAID or coxib use. Gastroenterology 2003, 124, 288-92. Lambert, M. P.; Barlow, A. K.; Chromy, B. A.; Edwards, C.; Freed, R.; Liosatos, M.; Morgan, T. E.; Rozovsky, I.; Trommer, B.; Viola, K. L.; Wals, P.; Zhang, C.; Finch, C. E.; Krafft, G. A.; Klein, W. L. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A 1998, 95, 6448-53. Lammich, S.; Kojro, E.; Postina, R.; Gilbert, S.; Pfeiffer, R.; Jasionowski, M.; Haass, C.; Fahrenholz, F. Constitutive and regulated alpha-secretase cleavage of Alzheimer's amyloid precursor protein by a disintegrin metalloprotease. Proc Natl Acad Sci U S A 1999, 96, 3922-7. Langman, M. J.; Morgan, L.; Worrall, A. Use of anti-inflammatory drugs by patients admitted with small or large bowel perforations and haemorrhage. Br Med J (Clin Res Ed) 1985, 290, 347-9. Launer, L. J.; Andersen, K.; Dewey, M. E.; Letenneur, L.; Ott, A.; Amaducci, L. A.; Brayne, C.; Copeland, J. R.; Dartigues, J. F.; Kragh-Sorensen, P.; Lobo, A.; Martinez-Lage, J. M.; Stijnen, T.; Hofman, A. Rates and risk factors for dementia and Alzheimer's disease: results from EURODEM pooled analyses. EURODEM Incidence Research Group and Work Groups. European Studies of Dementia. Neurology 1999, 52, 78-84. Le Bail, O.; Schmidt-Ullrich, R.; Israel, A. Promoter analysis of the gene encoding the I kappa Balpha/MAD3 inhibitor of NF-kappa B: positive regulation by members of the rel/NF-kappa B family. Embo J 1993, 12, 5043-9. Le Bars, P. L.; Katz, M. M.; Berman, N.; Itil, T. M.; Freedman, A. M.; Schatzberg, A. F. A placebocontrolled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. Jama 1997, 278, 1327-32. Le, Y.; Gong, W.; Tiffany, H. L.; Tumanov, A.; Nedospasov, S.; Shen, W.; Dunlop, N. M.; Gao, J. L.; Murphy, P. M.; Oppenheim, J. J.; Wang, J. M. Amyloid (beta)42 activates a G-protein-coupled chemoattractant receptor, FPR-like-1. J Neurosci 2001, 21, RC123. 70 REFERENCES L. Fisher LeClair, K. P.; Blanar, M. A.; Sharp, P. A. The p50 subunit of NF-kappa B associates with the NF-IL6 transcription factor. Proc Natl Acad Sci U S A 1992, 89, 8145-9. Lee, H. G.; Casadesus, G.; Zhu, X.; Takeda, A.; Perry, G.; Smith, M. A. Challenging the amyloid cascade hypothesis: senile plaques and amyloid-beta as protective adaptations to Alzheimer disease. Ann N Y Acad Sci 2004, 1019, 1-4. Lee, N. S.; Dohjima, T.; Bauer, G.; Li, H.; Li, M. J.; Ehsani, A.; Salvaterra, P.; Rossi, J. Expression of small interfering RNAs targeted against HIV-1 rev transcripts in human cells. Nat Biotechnol 2002, 20, 500-5. Lee, S. C.; Liu, W.; Dickson, D. W.; Brosnan, C. F.; Berman, J. W. Cytokine production by human fetal microglia and astrocytes. Differential induction by lipopolysaccharide and IL-1 beta. J Immunol 1993, 150, 2659-67. Lee, S. J.; Park, J. Y.; Hou, J.; Benveniste, E. N. Transcriptional regulation of the intercellular adhesion molecule-1 gene by proinflammatory cytokines in human astrocytes. Glia 1999, 25, 21-32. Lee, V. M. Biomedicine. Tauists and beta-aptists united--well almost! Science 2001, 293, 1446-7. Lee, V. M.; Balin, B. J.; Otvos, L., Jr.; Trojanowski, J. Q. A68: a major subunit of paired helical filaments and derivatized forms of normal Tau. Science 1991, 251, 675-8. Lee, Y. M.; Miau, L. H.; Chang, C. J.; Lee, S. C. Transcriptional induction of the alpha-1 acid glycoprotein (AGP) gene by synergistic interaction of two alternative activator forms of AGP/enhancer-binding protein (C/EBP beta) and NF-kappaB or Nopp140. Mol Cell Biol 1996, 16, 4257-63. Leifert, J. A.; Harkins, S.; Whitton, J. L. Full-length proteins attached to the HIV tat protein transduction domain are neither transduced between cells, nor exhibit enhanced immunogenicity. Gene Ther 2002, 9, 1422-8. Lesné, S.; Koh, M.T.; Kotilinek, L.; Kayed, R.; Glabe, C.G.; Yang, A.; Gallagher, M.; Ashe, K.H. A specific amyloid-β protein assembly in the brain impairs memory. Nature 2006, 440, 352-7. Levi-Strauss, M.; Mallat, M. Primary cultures of murine astrocytes produce C3 and factor B, two components of the alternative pathway of complement activation. J Immunol 1987, 139, 2361-6. Levy-Lahad, E.; Wasco, W.; Poorkaj, P.; Romano, D. M.; Oshima, J.; Pettingell, W. H.; Yu, C. E.; Jondro, P. D.; Schmidt, S. D.; Wang, K.; et al. Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science 1995, 269, 973-7. Lewis, J.; Dickson, D. W.; Lin, W. L.; Chisholm, L.; Corral, A.; Jones, G.; Yen, S. H.; Sahara, N.; Skipper, L.; Yager, D.; Eckman, C.; Hardy, J.; Hutton, M.; McGowan, E. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001, 293, 1487-91. Lewis, J.; McGowan, E.; Rockwood, J.; Melrose, H.; Nacharaju, P.; Van Slegtenhorst, M.; Gwinn-Hardy, K.; Paul Murphy, M.; Baker, M.; Yu, X.; Duff, K.; Hardy, J.; Corral, A.; Lin, W. L.; Yen, S. H.; Dickson, D. W.; Davies, P.; Hutton, M. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 2000, 25, 402-5. Lezoualc'h, F.; Sagara, Y.; Holsboer, F.; Behl, C. High constitutive NF-kappaB activity mediates resistance to oxidative stress in neuronal cells. J Neurosci 1998, 18, 3224-32. Li, Q.; Van Antwerp, D.; Mercurio, F.; Lee, K. F.; Verma, I. M. Severe liver degeneration in mice lacking the IkappaB kinase 2 gene. Science 1999, 284, 321-5. Li, Q. Q.; Bever, C. T.; Burt, D. R.; Judge, S. I.; Trisler, G. D. Induction of RANTES chemokine expression in human astrocytic cells is dependent upon activation of NF-kappaB transcription factor. Int J Mol Med 2001, 7, 527-33. Li, R.; Strohmeyer, R.; Liang, Z.; Lue, L. F.; Rogers, J. CCAAT/enhancer binding protein delta (C/EBPdelta) expression and elevation in Alzheimer's disease. Neurobiol Aging 2004, 25, 991-9. Li, T.; Ma, G.; Cai, H.; Price, D. L.; Wong, P. C. Nicastrin is required for assembly of presenilin/gammasecretase complexes to mediate Notch signaling and for processing and trafficking of beta-amyloid precursor protein in mammals. J Neurosci 2003, 23, 3272-7. Li, X.; Liao, W. S. Cooperative effects of C/EBP-like and NF kappa B-like binding sites on rat serum amyloid A1 gene expression in liver cells. Nucleic Acids Res 1992, 20, 4765-72. Li, Y.; Barger, S. W.; Liu, L.; Mrak, R. E.; Griffin, W. S. S100beta induction of the proinflammatory cytokine interleukin-6 in neurons. J Neurochem 2000, 74, 143-50. Libermann, T. A.; Baltimore, D. Activation of interleukin-6 gene expression through the NF-kappa B transcription factor. Mol Cell Biol 1990, 10, 2327-34. REFERENCES 71 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Licastro, F.; Grimaldi, L. M.; Bonafe, M.; Martina, C.; Olivieri, F.; Cavallone, L.; Giovanietti, S.; Masliah, E.; Franceschi, C. Interleukin-6 gene alleles affect the risk of Alzheimer's disease and levels of the cytokine in blood and brain. Neurobiol Aging 2003, 24, 921-6. Licastro, F.; Veglia, F.; Chiappelli, M.; Grimaldi, L. M.; Masliah, E. A polymorphism of the interleukin-1 beta gene at position +3953 influences progression and neuro-pathological hallmarks of Alzheimer's disease. Neurobiol Aging 2004, 25, 1017-22. Lieb, K.; Kaltschmidt, C.; Kaltschmidt, B.; Baeuerle, P. A.; Berger, M.; Bauer, J.; Fiebich, B. L. Interleukin-1 beta uses common and distinct signaling pathways for induction of the interleukin-6 and tumor necrosis factor alpha genes in the human astrocytoma cell line U373. J Neurochem 1996, 66, 1496-503. Liesi, P.; Silver, J. Is astrocyte laminin involved in axon guidance in the mammalian CNS? Dev Biol 1988, 130, 774-85. Lillien, L. E.; Raff, M. C. Differentiation signals in the CNS: type-2 astrocyte development in vitro as a model system. Neuron 1990, 5, 111-9. Lin, R.; Gewert, D.; Hiscott, J. Differential transcriptional activation in vitro by NF-kappa B/Rel proteins. J Biol Chem 1995, 270, 3123-31. Lindgren, M.; Hallbrink, M.; Prochiantz, A.; Langel, Ü. Cell-penetrating peptides. Trends Pharmacol Sci 2000, 21, 99-103. Lindgren, M. E.; Hallbrink, M. M.; Elmquist, A. M.; Langel, Ü. Passage of cell-penetrating peptides across a human epithelial cell layer in vitro. Biochem J 2004, 377, 69-76. Lindholm, D.; Heumann, R.; Meyer, M.; Thoenen, H. Interleukin-1 regulates synthesis of nerve growth factor in non-neuronal cells of rat sciatic nerve. Nature 1987, 330, 658-9. Ling, E. A.; Wong, W. C. The origin and nature of ramified and amoeboid microglia: a historical review and current concepts. Glia 1993, 7, 9-18. Lippa, C. F.; Nee, L. E.; Mori, H.; St George-Hyslop, P. Abeta-42 deposition precedes other changes in PS1 Alzheimer's disease. Lancet 1998, 352, 1117-8. Lipton, S. A. Janus faces of NF-kappa B: neurodestruction versus neuroprotection. Nat Med 1997, 3, 20-2. Liu, Q.; Kawai, H.; Berg, D. K. beta -Amyloid peptide blocks the response of alpha 7-containing nicotinic receptors on hippocampal neurons. Proc Natl Acad Sci U S A 2001, 98, 4734-9. Liu, Y.; Dargusch, R.; Schubert, D. Beta amyloid toxicity does not require RAGE protein. Biochem Biophys Res Commun 1997, 237, 37-40. Lorenzo, A.; Yankner, B. A. Beta-amyloid neurotoxicity requires fibril formation and is inhibited by congo red. Proc Natl Acad Sci U S A 1994, 91, 12243-7. Lovestone, S.; Reynolds, C. H. The phosphorylation of tau: a critical stage in neurodevelopment and neurodegenerative processes. Neuroscience 1997, 78, 309-24. Lue, L. F.; Brachova, L.; Civin, W. H.; Rogers, J. Inflammation, A beta deposition, and neurofibrillary tangle formation as correlates of Alzheimer's disease neurodegeneration. J Neuropathol Exp Neurol 1996, 55, 1083-8. Lue, L. F.; Kuo, Y. M.; Roher, A. E.; Brachova, L.; Shen, Y.; Sue, L.; Beach, T.; Kurth, J. H.; Rydel, R. E.; Rogers, J. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease. Am J Pathol 1999, 155, 853-62. Lue, L. F.; Walker, D. G.; Brachova, L.; Beach, T. G.; Rogers, J.; Schmidt, A. M.; Stern, D. M.; Yan, S. D. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer's disease: identification of a cellular activation mechanism. Exp Neurol 2001, 171, 29-45. Lundberg, M.; Johansson, M. Is VP22 nuclear homing an artifact? Nat Biotechnol 2001, 19, 713-4. Lundberg, M.; Johansson, M. Positively charged DNA-binding proteins cause apparent cell membrane translocation. Biochem Biophys Res Commun 2002, 291, 367-71. Luo, W. J.; Wang, H.; Li, H.; Kim, B. S.; Shah, S.; Lee, H. J.; Thinakaran, G.; Kim, T. W.; Yu, G.; Xu, H. PEN-2 and APH-1 coordinately regulate proteolytic processing of presenilin 1. J Biol Chem 2003, 278, 7850-4. Luo, Y.; Hattori, A.; Munoz, J.; Qin, Z. H.; Roth, G. S. Intrastriatal dopamine injection induces apoptosis through oxidation-involved activation of transcription factors AP-1 and NF-kappaB in rats. Mol Pharmacol 1999, 56, 254-64. Lust, J. A.; Donovan, K. A.; Kline, M. P.; Greipp, P. R.; Kyle, R. A.; Maihle, N. J. Isolation of an mRNA encoding a soluble form of the human interleukin-6 receptor. Cytokine 1992, 4, 96-100. 72 REFERENCES L. Fisher Mackenzie, I. R.; Hao, C.; Munoz, D. G. Role of microglia in senile plaque formation. Neurobiol Aging 1995, 16, 797-804. Magalini, A.; Savoldi, G.; Ferrari, F.; Garnier, M.; Ghezzi, P.; Albertini, A.; Di Lorenzo, D. Role of IL-1 beta and corticosteroids in the regulation of the C/EBP-alpha, beta and delta genes in vivo. Cytokine 1995, 7, 753-8. Maggirwar, S. B.; Sarmiere, P. D.; Dewhurst, S.; Freeman, R. S. Nerve growth factor-dependent activation of NF-kappaB contributes to survival of sympathetic neurons. J Neurosci 1998, 18, 10356-65. Majocha, R. E.; Agrawal, S.; Tang, J. Y.; Humke, E. W.; Marotta, C. A. Modulation of the PC12 cell response to nerve growth factor by antisense oligonucleotide to amyloid precursor protein. Cell Mol Neurobiol 1994, 14, 425-37. Mallat, M.; Houlgatte, R.; Brachet, P.; Prochiantz, A. Lipopolysaccharide-stimulated rat brain macrophages release NGF in vitro. Dev Biol 1989, 133, 309-11. Mann, M. J.; Dzau, V. J. Therapeutic applications of transcription factor decoy oligonucleotides. J Clin Invest 2000, 106, 1071-5. March, C. J.; Mosley, B.; Larsen, A.; Cerretti, D. P.; Braedt, G.; Price, V.; Gillis, S.; Henney, C. S.; Kronheim, S. R.; Grabstein, K.; et al. Cloning, sequence and expression of two distinct human interleukin-1 complementary DNAs. Nature 1985, 315, 641-7. Marchesi, V. T. An alternative interpretation of the amyloid Abeta hypothesis with regard to the pathogenesis of Alzheimer's disease. Proc Natl Acad Sci U S A 2005, 102, 9093-8. Marcus, D. L.; Thomas, C.; Rodriguez, C.; Simberkoff, K.; Tsai, J. S.; Strafaci, J. A.; Freedman, M. L. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer's disease. Exp Neurol 1998, 150, 40-4. Marz, P.; Cheng, J. G.; Gadient, R. A.; Patterson, P. H.; Stoyan, T.; Otten, U.; Rose-John, S. Sympathetic neurons can produce and respond to interleukin 6. Proc Natl Acad Sci U S A 1998, 95, 3251-6. Masters, C. L.; Simms, G.; Weinman, N. A.; Multhaup, G.; McDonald, B. L.; Beyreuther, K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci U S A 1985, 82, 4245-9. Matsusaka, T.; Fujikawa, K.; Nishio, Y.; Mukaida, N.; Matsushima, K.; Kishimoto, T.; Akira, S. Transcription factors NF-IL6 and NF-kappa B synergistically activate transcription of the inflammatory cytokines, interleukin 6 and interleukin 8. Proc Natl Acad Sci U S A 1993, 90, 10193-7. Matsuzaki, K. Magainins as paradigm for the mode of action of pore forming polypeptides. Biochim Biophys Acta 1998, 1376, 391-400. Matsuzaki, K.; Yoneyama, S.; Miyajima, K. Pore formation and translocation of melittin. Biophys J 1997, 73, 831-8. Matsuzaki, K.; Yoneyama, S.; Murase, O.; Miyajima, K. Transbilayer transport of ions and lipids coupled with mastoparan X translocation. Biochemistry 1996, 35, 8450-6. Mattson, M. P.; Cheng, B.; Davis, D.; Bryant, K.; Lieberburg, I.; Rydel, R. E. beta-Amyloid peptides destabilize calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity. J Neurosci 1992, 12, 376-89. Mattson, M. P.; Culmsee, C.; Yu, Z.; Camandola, S. Roles of nuclear factor kappaB in neuronal survival and plasticity. J Neurochem 2000, 74, 443-56. Mattson, M. P.; Goodman, Y.; Luo, H.; Fu, W.; Furukawa, K. Activation of NF-kappaB protects hippocampal neurons against oxidative stress-induced apoptosis: evidence for induction of manganese superoxide dismutase and suppression of peroxynitrite production and protein tyrosine nitration. J Neurosci Res 1997, 49, 681-97. McGeer, E. G.; McGeer, P. L. The importance of inflammatory mechanisms in Alzheimer disease. Exp Gerontol 1998, 33, 371-8. McGeer, E. G.; McGeer, P. L. Inflammatory processes in Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2003, 27, 741-9. McGeer, P. L.; Akiyama, H.; Itagaki, S.; McGeer, E. G. Activation of the classical complement pathway in brain tissue of Alzheimer patients. Neurosci Lett 1989, 107, 341-6. McGeer, P. L.; Itagaki, S.; McGeer, E. G. Expression of the histocompatibility glycoprotein HLA-DR in neurological disease. Acta Neuropathol (Berl) 1988, 76, 550-7. REFERENCES 73 Inflammatory Cytokines and NFκB in Alzheimer’s Disease McGeer, P. L.; Schulzer, M.; McGeer, E. G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer's disease: a review of 17 epidemiologic studies. Neurology 1996, 47, 42532. McIntyre, K. W.; Shuster, D. J.; Gillooly, K. M.; Dambach, D. M.; Pattoli, M. A.; Lu, P.; Zhou, X. D.; Qiu, Y.; Zusi, F. C.; Burke, J. R. A highly selective inhibitor of I kappa B kinase, BMS-345541, blocks both joint inflammation and destruction in collagen-induced arthritis in mice. Arthritis Rheum 2003, 48, 2652-9. McKee, A. C.; Kowall, N. W.; Schumacher, J. S.; Beal, M. F. The neurotoxicity of amyloid beta protein in aged primates. Amyloid 1998, 5, 1-9. McMahan, C. J.; Slack, J. L.; Mosley, B.; Cosman, D.; Lupton, S. D.; Brunton, L. L.; Grubin, C. E.; Wignall, J. M.; Jenkins, N. A.; Brannan, C. I.; et al. A novel IL-1 receptor, cloned from B cells by mammalian expression, is expressed in many cell types. Embo J 1991, 10, 2821-32. McMahon, B. M.; Stewart, J.; Fauq, A.; Younkin, S.; Younkin, L.; Richelson, E. Using peptide nucleic acids as gene-expression modifiers to reduce beta-amyloid levels. J Mol Neurosci 2002a, 19, 71-6. McMahon, B. M.; Stewart, J. A.; Bitner, M. D.; Fauq, A.; McCormick, D. J.; Richelson, E. Peptide nucleic acids specifically cause antigene effects in vivo by systemic injection. Life Sci 2002b, 71, 325-37. Mecocci, P.; MacGarvey, U.; Beal, M. F. Oxidative damage to mitochondrial DNA is increased in Alzheimer's disease. Ann Neurol 1994, 36, 747-51. Merola, M.; Blanchard, B.; Tovey, M. G. The kappa B enhancer of the human interleukin-6 promoter is necessary and sufficient to confer an IL-1 beta and TNF-alpha response in transfected human cell lines: requirement for members of the C/EBP family for activity. J Interferon Cytokine Res 1996, 16, 783-98. Merrill, J. E. Effects of interleukin-1 and tumor necrosis factor-alpha on astrocytes, microglia, oligodendrocytes, and glial precursors in vitro. Dev Neurosci 1991, 13, 130-7. Middleton, G.; Hamanoue, M.; Enokido, Y.; Wyatt, S.; Pennica, D.; Jaffray, E.; Hay, R. T.; Davies, A. M. Cytokine-induced nuclear factor kappa B activation promotes the survival of developing neurons. J Cell Biol 2000, 148, 325-32. Mitchell, D. J.; Kim, D. T.; Steinman, L.; Fathman, C. G.; Rothbard, J. B. Polyarginine enters cells more efficiently than other polycationic homopolymers. J Pept Res 2000, 56, 318-25. Mitrasinovic, O. M.; Murphy, G. M., Jr. Microglial overexpression of the M-CSF receptor augments phagocytosis of opsonized Abeta. Neurobiol Aging 2003, 24, 807-15. Miyazawa, K.; Mori, A.; Yamamoto, K.; Okudaira, H. Transcriptional roles of CCAAT/enhancer binding protein-beta, nuclear factor-kappaB, and C-promoter binding factor 1 in interleukin (IL)-1betainduced IL-6 synthesis by human rheumatoid fibroblast-like synoviocytes. J Biol Chem 1998, 273, 7620-7. Mochida, Y.; Takeda, K.; Saitoh, M.; Nishitoh, H.; Amagasa, T.; Ninomiya-Tsuji, J.; Matsumoto, K.; Ichijo, H. ASK1 inhibits interleukin-1-induced NF-kappa B activity through disruption of TRAF6TAK1 interaction. J Biol Chem 2000, 275, 32747-52. Moerman, A. M.; Mao, X.; Lucas, M. M.; Barger, S. W. Characterization of a neuronal kappaB-binding factor distinct from NF-kappaB. Brain Res Mol Brain Res 1999, 67, 303-15. Mogensen, H. S.; Beatty, D. M.; Morris, S. J.; Jorgensen, O. S. Amyloid beta-peptide(25-35) changes [Ca2+] in hippocampal neurons. Neuroreport 1998, 9, 1553-8. Morishita, R.; Higaki, J.; Tomita, N.; Ogihara, T. Application of transcription factor "decoy" strategy as means of gene therapy and study of gene expression in cardiovascular disease. Circ Res 1998, 82, 1023-8. Morishita, R.; Sugimoto, T.; Aoki, M.; Kida, I.; Tomita, N.; Moriguchi, A.; Maeda, K.; Sawa, Y.; Kaneda, Y.; Higaki, J.; Ogihara, T. In vivo transfection of cis element "decoy" against nuclear factorkappaB binding site prevents myocardial infarction. Nat Med 1997, 3, 894-9. Morris, J. C.; Storandt, M.; McKeel, D. W., Jr.; Rubin, E. H.; Price, J. L.; Grant, E. A.; Berg, L. Cerebral amyloid deposition and diffuse plaques in "normal" aging: Evidence for presymptomatic and very mild Alzheimer's disease. Neurology 1996, 46, 707-19. Mortimer, J. A.; van Duijn, C. M.; Chandra, V.; Fratiglioni, L.; Graves, A. B.; Heyman, A.; Jorm, A. F.; Kokmen, E.; Kondo, K.; Rocca, W. A.; et al. Head trauma as a risk factor for Alzheimer's disease: a collaborative re-analysis of case-control studies. EURODEM Risk Factors Research Group. Int J Epidemiol 1991, 20 Suppl 2, S28-35. 74 REFERENCES L. Fisher Moynagh, P. N.; Williams, D. C.; O'Neill, L. A. Interleukin-1 activates transcription factor NF kappa B in glial cells. Biochem J 1993, 294 ( Pt 2), 343-7. Mucke, L.; Yu, G. Q.; McConlogue, L.; Rockenstein, E. M.; Abraham, C. R.; Masliah, E. Astroglial expression of human alpha(1)-antichymotrypsin enhances alzheimer-like pathology in amyloid protein precursor transgenic mice. Am J Pathol 2000, 157, 2003-10. Mullan, M.; Crawford, F.; Axelman, K.; Houlden, H.; Lilius, L.; Winblad, B.; Lannfelt, L. A pathogenic mutation for probable Alzheimer's disease in the APP gene at the N-terminus of beta-amyloid. Nat Genet 1992, 1, 345-7. Mullberg, J.; Schooltink, H.; Stoyan, T.; Gunther, M.; Graeve, L.; Buse, G.; Mackiewicz, A.; Heinrich, P. C.; Rose-John, S. The soluble interleukin-6 receptor is generated by shedding. Eur J Immunol 1993, 23, 473-80. Muller, J. M.; Rupec, R. A.; Baeuerle, P. A. Study of gene regulation by NF-kappa B and AP-1 in response to reactive oxygen intermediates. Methods 1997, 11, 301-12. Murphy, G. M., Jr.; Yang, L.; Cordell, B. Macrophage colony-stimulating factor augments beta-amyloidinduced interleukin-1, interleukin-6, and nitric oxide production by microglial cells. J Biol Chem 1998, 273, 20967-71. Murrell, J.; Farlow, M.; Ghetti, B.; Benson, M. D. A mutation in the amyloid precursor protein associated with hereditary Alzheimer's disease. Science 1991, 254, 97-9. Nagele, R. G.; D'Andrea, M. R.; Lee, H.; Venkataraman, V.; Wang, H. Y. Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res 2003, 971, 197209. Naslund, J.; Haroutunian, V.; Mohs, R.; Davis, K. L.; Davies, P.; Greengard, P.; Buxbaum, J. D. Correlation between elevated levels of amyloid beta-peptide in the brain and cognitive decline. Jama 2000, 283, 1571-7. Natsuka, S.; Akira, S.; Nishio, Y.; Hashimoto, S.; Sugita, T.; Isshiki, H.; Kishimoto, T. Macrophage differentiation-specific expression of NF-IL6, a transcription factor for interleukin-6. Blood 1992, 79, 460-6. Neeper, M.; Schmidt, A. M.; Brett, J.; Yan, S. D.; Wang, F.; Pan, Y. C.; Elliston, K.; Stern, D.; Shaw, A. Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J Biol Chem 1992, 267, 14998-5004. Nekhotiaeva, N.; Elmquist, A.; Rajarao, G. K.; Hallbrink, M.; Langel, U.; Good, L. Cell entry and antimicrobial properties of eukaryotic cell-penetrating peptides. Faseb J 2004, 18, 394-6. Ng, S. B.; Tan, Y. H.; Guy, G. R. Differential induction of the interleukin-6 gene by tumor necrosis factor and interleukin-1. J Biol Chem 1994, 269, 19021-7. Nicoll, J. A.; Mrak, R. E.; Graham, D. I.; Stewart, J.; Wilcock, G.; MacGowan, S.; Esiri, M. M.; Murray, L. S.; Dewar, D.; Love, S.; Moss, T.; Griffin, W. S. Association of interleukin-1 gene polymorphisms with Alzheimer's disease. Ann Neurol 2000, 47, 365-8. Nielsen, P. E.; Egholm, M.; Berg, R. H.; Buchardt, O. Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide. Science 1991, 254, 1497-500. Nielsen, P. E.; Egholm, M.; Berg, R. H.; Buchardt, O. Peptide nucleic acids (PNAs): potential antisense and anti-gene agents. Anticancer Drug Des 1993, 8, 53-63. Nilsberth, C.; Westlind-Danielsson, A.; Eckman, C. B.; Condron, M. M.; Axelman, K.; Forsell, C.; Stenh, C.; Luthman, J.; Teplow, D. B.; Younkin, S. G.; Naslund, J.; Lannfelt, L. The 'Arctic' APP mutation (E693G) causes Alzheimer's disease by enhanced Abeta protofibril formation. Nat Neurosci 2001, 4, 887-93. Nilsson, L. N.; Bales, K. R.; DiCarlo, G.; Gordon, M. N.; Morgan, D.; Paul, S. M.; Potter, H. Alpha-1antichymotrypsin promotes beta-sheet amyloid plaque deposition in a transgenic mouse model of Alzheimer's disease. J Neurosci 2001, 21, 1444-51. Nurmi, A.; Vartiainen, N.; Pihlaja, R.; Goldsteins, G.; Yrjanheikki, J.; Koistinaho, J. Pyrrolidine dithiocarbamate inhibits translocation of nuclear factor kappa-B in neurons and protects against brain ischaemia with a wide therapeutic time window. J Neurochem 2004, 91, 755-65. O'Barr, S.; Cooper, N. R. The C5a complement activation peptide increases IL-1beta and IL-6 release from amyloid-beta primed human monocytes: implications for Alzheimer's disease. J Neuroimmunol 2000, 109, 87-94. REFERENCES 75 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Oddo, S.; Caccamo, A.; Shepherd, J. D.; Murphy, M. P.; Golde, T. E.; Kayed, R.; Metherate, R.; Mattson, M. P.; Akbari, Y.; LaFerla, F. M. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 2003, 39, 409-21. Oitzl, M. S.; van Oers, H.; Schobitz, B.; de Kloet, E. R. Interleukin-1 beta, but not interleukin-6, impairs spatial navigation learning. Brain Res 1993, 613, 160-3. Olson, M. I.; Shaw, C. M. Presenile dementia and Alzheimer's disease in mongolism. Brain 1969, 92, 14756. O'Neill, L. A.; Kaltschmidt, C. NF-kappa B: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci 1997, 20, 252-8. Ostenson, C. G.; Sandberg-Nordqvist, A. C.; Chen, J.; Hallbrink, M.; Rotin, D.; Langel, U.; Efendic, S. Overexpression of protein-tyrosine phosphatase PTP sigma is linked to impaired glucose-induced insulin secretion in hereditary diabetic Goto-Kakizaki rats. Biochem Biophys Res Commun 2002, 291, 945-50. Packer, L.; Suzuki, Y. J. Vitamin E and alpha-lipoate: role in antioxidant recycling and activation of the NF-kappa B transcription factor. Mol Aspects Med 1993, 14, 229-39. Pahl, H. L. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene 1999, 18, 685366. Palmer, A. M.; Burns, M. A. Selective increase in lipid peroxidation in the inferior temporal cortex in Alzheimer's disease. Brain Res 1994, 645, 338-42. Palmert, M. R.; Podlisny, M. B.; Witker, D. S.; Oltersdorf, T.; Younkin, L. H.; Selkoe, D. J.; Younkin, S. G. The beta-amyloid protein precursor of Alzheimer disease has soluble derivatives found in human brain and cerebrospinal fluid. Proc Natl Acad Sci U S A 1989, 86, 6338-42. Panegyres, P. K. The functions of the amyloid precursor protein gene. Rev Neurosci 2001, 12, 1-39. Papassotiropoulos, A.; Bagli, M.; Jessen, F.; Bayer, T. A.; Maier, W.; Rao, M. L.; Heun, R. A genetic variation of the inflammatory cytokine interleukin-6 delays the initial onset and reduces the risk for sporadic Alzheimer's disease. Ann Neurol 1999, 45, 666-8. Paresce, D. M.; Chung, H.; Maxfield, F. R. Slow degradation of aggregates of the Alzheimer's disease amyloid beta-protein by microglial cells. J Biol Chem 1997, 272, 29390-7. Parry, G. C.; Mackman, N. A set of inducible genes expressed by activated human monocytic and endothelial cells contain kappa B-like sites that specifically bind c-Rel-p65 heterodimers. J Biol Chem 1994, 269, 20823-5. Pasinetti, G. M. From epidemiology to therapeutic trials with anti-inflammatory drugs in Alzheimer's disease: the role of NSAIDs and cyclooxygenase in beta-amyloidosis and clinical dementia. J Alzheimers Dis 2002, 4, 435-45. Peacock, M. L.; Warren, J. T., Jr.; Roses, A. D.; Fink, J. K. Novel polymorphism in the A4 region of the amyloid precursor protein gene in a patient without Alzheimer's disease. Neurology 1993, 43, 1254-6. Perkins, N. D. Achieving transcriptional specificity with NF-kappa B. Int J Biochem Cell Biol 1997, 29, 1433-48. Perkins, N. D.; Agranoff, A. B.; Pascal, E.; Nabel, G. J. An interaction between the DNA-binding domains of RelA(p65) and Sp1 mediates human immunodeficiency virus gene activation. Mol Cell Biol 1994, 14, 6570-83. Perkins, N. D.; Edwards, N. L.; Duckett, C. S.; Agranoff, A. B.; Schmid, R. M.; Nabel, G. J. A cooperative interaction between NF-kappa B and Sp1 is required for HIV-1 enhancer activation. Embo J 1993, 12, 3551-8. Perry, V. H.; Newman, T. A.; Cunningham, C. The impact of systemic infection on the progression of neurodegenerative disease. Nat Rev Neurosci 2003, 4, 103-12. Pichon, C.; Monsigny, M.; Roche, A. C. Intracellular localization of oligonucleotides: influence of fixative protocols. Antisense Nucleic Acid Drug Dev 1999, 9, 89-93. Pierce, J. W.; Read, M. A.; Ding, H.; Luscinskas, F. W.; Collins, T. Salicylates inhibit I kappa B-alpha phosphorylation, endothelial-leukocyte adhesion molecule expression, and neutrophil transmigration. J Immunol 1996, 156, 3961-9. Pike, C. J.; Burdick, D.; Walencewicz, A. J.; Glabe, C. G.; Cotman, C. W. Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J Neurosci 1993, 13, 1676-87. Pike, C. J.; Cummings, B. J.; Cotman, C. W. beta-Amyloid induces neuritic dystrophy in vitro: similarities with Alzheimer pathology. Neuroreport 1992, 3, 769-72. 76 REFERENCES L. Fisher Pike, C. J.; Cummings, B. J.; Cotman, C. W. Early association of reactive astrocytes with senile plaques in Alzheimer's disease. Exp Neurol 1995a, 132, 172-9. Pike, C. J.; Cummings, B. J.; Monzavi, R.; Cotman, C. W. Beta-amyloid-induced changes in cultured astrocytes parallel reactive astrocytosis associated with senile plaques in Alzheimer's disease. Neuroscience 1994, 63, 517-31. Pike, C. J.; Walencewicz, A. J.; Glabe, C. G.; Cotman, C. W. Aggregation-related toxicity of synthetic beta-amyloid protein in hippocampal cultures. Eur J Pharmacol 1991a, 207, 367-8. Pike, C. J.; Walencewicz, A. J.; Glabe, C. G.; Cotman, C. W. In vitro aging of beta-amyloid protein causes peptide aggregation and neurotoxicity. Brain Res 1991b, 563, 311-4. Pike, C. J.; Walencewicz-Wasserman, A. J.; Kosmoski, J.; Cribbs, D. H.; Glabe, C. G.; Cotman, C. W. Structure-activity analyses of beta-amyloid peptides: contributions of the beta 25-35 region to aggregation and neurotoxicity. J Neurochem 1995b, 64, 253-65. Pinnix, I.; Musunuru, U.; Tun, H.; Sridharan, A.; Golde, T.; Eckman, C.; Ziani-Cherif, C.; Onstead, L.; Sambamurti, K. A novel gamma -secretase assay based on detection of the putative C-terminal fragment-gamma of amyloid beta protein precursor. J Biol Chem 2001, 276, 481-7. Pizzi, M.; Goffi, F.; Boroni, F.; Benarese, M.; Perkins, S. E.; Liou, H. C.; Spano, P. Opposing roles for NFkappa B/Rel factors p65 and c-Rel in the modulation of neuron survival elicited by glutamate and interleukin-1beta. J Biol Chem 2002, 277, 20717-23. Plevy, S. E.; Gemberling, J. H.; Hsu, S.; Dorner, A. J.; Smale, S. T. Multiple control elements mediate activation of the murine and human interleukin 12 p40 promoters: evidence of functional synergy between C/EBP and Rel proteins. Mol Cell Biol 1997, 17, 4572-88. Pola, R.; Flex, A.; Gaetani, E.; Lago, A. D.; Gerardino, L.; Pola, P.; Bernabei, R. The -174 G/C polymorphism of the interleukin-6 gene promoter is associated with Alzheimer's disease in an Italian population [corrected]. Neuroreport 2002, 13, 1645-7. Ponte, P.; Gonzalez-DeWhitt, P.; Schilling, J.; Miller, J.; Hsu, D.; Greenberg, B.; Davis, K.; Wallace, W.; Lieberburg, I.; Fuller, F. A new A4 amyloid mRNA contains a domain homologous to serine proteinase inhibitors. Nature 1988, 331, 525-7. Pooga, M.; Hallbrink, M.; Zorko, M.; Langel, Ü. Cell penetration by transportan. Faseb J 1998a, 12, 67-77. Pooga, M.; Kut, C.; Kihlmark, M.; Hallbrink, M.; Fernaeus, S.; Raid, R.; Land, T.; Hallberg, E.; Bartfai, T.; Langel, Ü. Cellular translocation of proteins by transportan. Faseb J 2001, 15, 1451-3. Pooga, M.; Soomets, U.; Hallbrink, M.; Valkna, A.; Saar, K.; Rezaei, K.; Kahl, U.; Hao, J. X.; Xu, X. J.; Wiesenfeld-Hallin, Z.; Hokfelt, T.; Bartfai, T.; Langel, Ü. Cell penetrating PNA constructs regulate galanin receptor levels and modify pain transmission in vivo. Nat Biotechnol 1998b, 16, 857-61. Poon, H. F.; Joshi, G.; Sultana, R.; Farr, S. A.; Banks, W. A.; Morley, J. E.; Calabrese, V.; Butterfield, D. A. Antisense directed at the Abeta region of APP decreases brain oxidative markers in aged senescence accelerated mice. Brain Res 2004, 1018, 86-96. Post, A.; Holsboer, F.; Behl, C. Induction of NF-kappaB activity during haloperidol-induced oxidative toxicity in clonal hippocampal cells: suppression of NF-kappaB and neuroprotection by antioxidants. J Neurosci 1998, 18, 8236-46. Pratico, D.; Delanty, N. Oxidative injury in diseases of the central nervous system: focus on Alzheimer's disease. Am J Med 2000, 109, 577-85. Prechel, M. M.; Halbur, L.; Devata, S.; Vaidya, A. M.; Young, M. R. Increased interleukin-6 production by cerebral cortical tissue of adult versus young mice. Mech Ageing Dev 1996, 92, 185-94. Puglielli, L.; Friedlich, A.L.; Setchell, K.D.; Nagano, S.; Opazo, C.; Cherny, R.A.; Barnham, K.J.; Wade, J.D.; Melov, S.; Kovacs, D.M.; Bush, A.I. Alzheimer disease amyloid-beta activity mimics cholesterol oxidase. J Clin Invest 2005, 115, 2556-63. Qin, Z. H.; Wang, Y.; Nakai, M.; Chase, T. N. Nuclear factor-kappa B contributes to excitotoxin-induced apoptosis in rat striatum. Mol Pharmacol 1998, 53, 33-42. Qiu, Z.; Sweeney, D. D.; Netzeband, J. G.; Gruol, D. L. Chronic interleukin-6 alters NMDA receptormediated membrane responses and enhances neurotoxicity in developing CNS neurons. J Neurosci 1998, 18, 10445-56. Quintanilla, R.A.; Orellana, D.I.; González-Billault, C.; Maccioni, R.B. Interleukin-6 induces Alzheimertype phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res 2004, 295, 245-57. Raff, M. C. Glial cell diversification in the rat optic nerve. Science 1989, 243, 1450-5. REFERENCES 77 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Raff, M. C.; Miller, R. H.; Noble, M. A glial progenitor cell that develops in vitro into an astrocyte or an oligodendrocyte depending on culture medium. Nature 1983, 303, 390-6. Rank, K. B.; Pauley, A. M.; Bhattacharya, K.; Wang, Z.; Evans, D. B.; Fleck, T. J.; Johnston, J. A.; Sharma, S. K. Direct interaction of soluble human recombinant tau protein with Abeta 1-42 results in tau aggregation and hyperphosphorylation by tau protein kinase II. FEBS Lett 2002, 514, 263-8. Ray, A.; Hannink, M.; Ray, B. K. Concerted participation of NF-kappa B and C/EBP heteromer in lipopolysaccharide induction of serum amyloid A gene expression in liver. J Biol Chem 1995, 270, 7365-74. Ray, A.; Prefontaine, K. E.; Ray, P. Down-modulation of interleukin-6 gene expression by 17 beta-estradiol in the absence of high affinity DNA binding by the estrogen receptor. J Biol Chem 1994, 269, 12940-6. Reier, P. J.; Houle, J. D. The glial scar: its bearing on axonal elongation and transplantation approaches to CNS repair. Adv Neurol 1988, 47, 87-138. Reines, S. A.; Block, G. A.; Morris, J. C.; Liu, G.; Nessly, M. L.; Lines, C. R.; Norman, B. A.; Baranak, C. C. Rofecoxib: no effect on Alzheimer's disease in a 1-year, randomized, blinded, controlled study. Neurology 2004, 62, 66-71. Rezaei, K.; Xu, I. S.; Wu, W. P.; Shi, T. J.; Soomets, U.; Land, T.; Xu, X. J.; Wiesenfeld-Hallin, Z.; Hokfelt, T.; Bartfai, T.; Langel, U. Intrathecal administration of PNA targeting galanin receptor reduces galanin-mediated inhibitory effect in the rat spinal cord. Neuroreport 2001, 12, 317-20. Ricciarelli, R.; D'Abramo, C.; Zingg, J. M.; Giliberto, L.; Markesbery, W.; Azzi, A.; Marinari, U. M.; Pronzato, M. A.; Tabaton, M. CD36 overexpression in human brain correlates with beta-amyloid deposition but not with Alzheimer's disease. Free Radic Biol Med 2004, 36, 1018-24. Richard, J. P.; Melikov, K.; Vives, E.; Ramos, C.; Verbeure, B.; Gait, M. J.; Chernomordik, L. V.; Lebleu, B. Cell-penetrating peptides. A reevaluation of the mechanism of cellular uptake. J Biol Chem 2003, 278, 585-90. Robakis, N. K.; Ramakrishna, N.; Wolfe, G.; Wisniewski, H. M. Molecular cloning and characterization of a cDNA encoding the cerebrovascular and the neuritic plaque amyloid peptides. Proc Natl Acad Sci U S A 1987, 84, 4190-4. Roberts, G. W.; Gentleman, S. M.; Lynch, A.; Graham, D. I. beta A4 amyloid protein deposition in brain after head trauma. Lancet 1991, 338, 1422-3. Rogaev, E. I.; Sherrington, R.; Rogaeva, E. A.; Levesque, G.; Ikeda, M.; Liang, Y.; Chi, H.; Lin, C.; Holman, K.; Tsuda, T.; et al. Familial Alzheimer's disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer's disease type 3 gene. Nature 1995, 376, 775-8. Rogers, J.; Cooper, N. R.; Webster, S.; Schultz, J.; McGeer, P. L.; Styren, S. D.; Civin, W. H.; Brachova, L.; Bradt, B.; Ward, P.; et al. Complement activation by beta-amyloid in Alzheimer disease. Proc Natl Acad Sci U S A 1992, 89, 10016-20. Rogers, J.; Kirby, L. C.; Hempelman, S. R.; Berry, D. L.; McGeer, P. L.; Kaszniak, A. W.; Zalinski, J.; Cofield, M.; Mansukhani, L.; Willson, P.; et al. Clinical trial of indomethacin in Alzheimer's disease. Neurology 1993, 43, 1609-11. Rogers, J. T.; Leiter, L. M.; McPhee, J.; Cahill, C. M.; Zhan, S. S.; Potter, H.; Nilsson, L. N. Translation of the alzheimer amyloid precursor protein mRNA is up-regulated by interleukin-1 through 5'untranslated region sequences. J Biol Chem 1999, 274, 6421-31. Roher, A. E.; Chaney, M. O.; Kuo, Y. M.; Webster, S. D.; Stine, W. B.; Haverkamp, L. J.; Woods, A. S.; Cotter, R. J.; Tuohy, J. M.; Krafft, G. A.; Bonnell, B. S.; Emmerling, M. R. Morphology and toxicity of Abeta-(1-42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer's disease. J Biol Chem 1996, 271, 20631-5. Roldan, A.; Gogg, S.; Ferrini, M.; Schillaci, R.; De Nicola, A. F. Glucocorticoid regulation of in vitro astrocyte phagocytosis. Biocell 1997, 21, 83-9. Roshak, A. K.; Callahan, J. F.; Blake, S. M. Small-molecule inhibitors of NF-kappaB for the treatment of inflammatory joint disease. Curr Opin Pharmacol 2002, 2, 316-21. Rossi, A.; Elia, G.; Santoro, M. G. Activation of the heat shock factor 1 by serine protease inhibitors. An effect associated with nuclear factor-kappaB inhibition. J Biol Chem 1998, 273, 16446-52. Rossi, A.; Kapahi, P.; Natoli, G.; Takahashi, T.; Chen, Y.; Karin, M.; Santoro, M. G. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IkappaB kinase. Nature 2000, 403, 103-8. Rothwell, N. J.; Luheshi, G.; Toulmond, S. Cytokines and their receptors in the central nervous system: physiology, pharmacology, and pathology. Pharmacol Ther 1996, 69, 85-95. 78 REFERENCES L. Fisher Rottkamp, C. A.; Raina, A. K.; Zhu, X.; Gaier, E.; Bush, A. I.; Atwood, C. S.; Chevion, M.; Perry, G.; Smith, M. A. Redox-active iron mediates amyloid-beta toxicity. Free Radic Biol Med 2001, 30, 447-50. Rush, D. K.; Aschmies, S.; Merriman, M. C. Intracerebral beta-amyloid(25-35) produces tissue damage: is it neurotoxic? Neurobiol Aging 1992, 13, 591-4. Russo-Marie, F. Macrophages and the glucocorticoids. J Neuroimmunol 1992, 40, 281-6. Saalik, P.; Elmquist, A.; Hansen, M.; Padari, K.; Saar, K.; Viht, K.; Langel, Ü.; Pooga, M. Protein cargo delivery properties of cell-penetrating peptides. A comparative study. Bioconjug Chem 2004, 15, 1246-53. Saar, K.; Lindgren, M.; Hansen, M.; Eiriksdottir, E.; Jiang, Y.; Rosenthal-Aizman, K.; Sassian, M.; Langel, Ü. Cell-penetrating peptides: a comparative membrane toxicity study. Anal Biochem 2005, 345, 55-65. Samuelsson, M.; Fisher, L.; Iverfeldt, K. beta-Amyloid and interleukin-1beta induce persistent NF-kappaB activation in rat primary glial cells. Int J Mol Med 2005, 16, 449-53. Sandler, S.; Bendtzen, K.; Eizirik, D. L.; Sjoholm, A.; Welsh, N. Decreased cell replication and polyamine content in insulin-producing cells after exposure to human interleukin 1 beta. Immunol Lett 1989, 22, 267-72. Sano, M.; Ernesto, C.; Thomas, R. G.; Klauber, M. R.; Schafer, K.; Grundman, M.; Woodbury, P.; Growdon, J.; Cotman, C. W.; Pfeiffer, E.; Schneider, L. S.; Thal, L. J. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. N Engl J Med 1997, 336, 1216-22. Sapolsky, R. M.; Uno, H.; Rebert, C. S.; Finch, C. E. Hippocampal damage associated with prolonged glucocorticoid exposure in primates. J Neurosci 1990, 10, 2897-902. Sasaki, A.; Yamaguchi, H.; Ogawa, A.; Sugihara, S.; Nakazato, Y. Microglial activation in early stages of amyloid beta protein deposition. Acta Neuropathol (Berl) 1997, 94, 316-22. Sasaki, N.; Toki, S.; Chowei, H.; Saito, T.; Nakano, N.; Hayashi, Y.; Takeuchi, M.; Makita, Z. Immunohistochemical distribution of the receptor for advanced glycation end products in neurons and astrocytes in Alzheimer's disease. Brain Res 2001, 888, 256-262. Sastre, M.; Steiner, H.; Fuchs, K.; Capell, A.; Multhaup, G.; Condron, M.M.; Teplow, D.B.; Haass, C. Presenilin-dependent γ-secretase processing of β-amyloid precursor protein at a site corresponding to the S3 cleavage of Notch. EMBO Rep 2001, 2, 835-42. Scheinman, R. I.; Gualberto, A.; Jewell, C. M.; Cidlowski, J. A.; Baldwin, A. S., Jr. Characterization of mechanisms involved in transrepression of NF-kappa B by activated glucocorticoid receptors. Mol Cell Biol 1995, 15, 943-53. Scherr, M.; Morgan, M. A.; Eder, M. Gene silencing mediated by small interfering RNAs in mammalian cells. Curr Med Chem 2003, 10, 245-56. Scheuner, D.; Eckman, C.; Jensen, M.; Song, X.; Citron, M.; Suzuki, N.; Bird, T. D.; Hardy, J.; Hutton, M.; Kukull, W.; Larson, E.; Levy-Lahad, E.; Viitanen, M.; Peskind, E.; Poorkaj, P.; Schellenberg, G.; Tanzi, R.; Wasco, W.; Lannfelt, L.; Selkoe, D.; Younkin, S. Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nat Med 1996, 2, 864-70. Schindler, R.; Gelfand, J. A.; Dinarello, C. A. Recombinant C5a stimulates transcription rather than translation of interleukin-1 (IL-1) and tumor necrosis factor: translational signal provided by lipopolysaccharide or IL-1 itself. Blood 1990, 76, 1631-8. Schmedtje, J. F., Jr.; Ji, Y. S.; Liu, W. L.; DuBois, R. N.; Runge, M. S. Hypoxia induces cyclooxygenase-2 via the NF-kappaB p65 transcription factor in human vascular endothelial cells. J Biol Chem 1997, 272, 601-8. Schmid, R. M.; Perkins, N. D.; Duckett, C. S.; Andrews, P. C.; Nabel, G. J. Cloning of an NF-kappa B subunit which stimulates HIV transcription in synergy with p65. Nature 1991, 352, 733-6. Schmidt, A. M.; Yan, S. D.; Yan, S. F.; Stern, D. M. The biology of the receptor for advanced glycation end products and its ligands. Biochim Biophys Acta 2000, 1498, 99-111. Schmitz, C.; Rutten, B. P.; Pielen, A.; Schafer, S.; Wirths, O.; Tremp, G.; Czech, C.; Blanchard, V.; Multhaup, G.; Rezaie, P.; Korr, H.; Steinbusch, H. W.; Pradier, L.; Bayer, T. A. Hippocampal neuron loss exceeds amyloid plaque load in a transgenic mouse model of Alzheimer's disease. Am J Pathol 2004, 164, 1495-502. REFERENCES 79 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Schneider, A.; Martin-Villalba, A.; Weih, F.; Vogel, J.; Wirth, T.; Schwaninger, M. NF-kappaB is activated and promotes cell death in focal cerebral ischemia. Nat Med 1999, 5, 554-9. Schreck, R.; Grassmann, R.; Fleckenstein, B.; Baeuerle, P. A. Antioxidants selectively suppress activation of NF-kappa B by human T-cell leukemia virus type I Tax protein. J Virol 1992, 66, 6288-93. Schwab, C.; Hosokawa, M.; McGeer, P. L. Transgenic mice overexpressing amyloid beta protein are an incomplete model of Alzheimer disease. Exp Neurol 2004, 188, 52-64. Sciacca, F. L.; Ferri, C.; Licastro, F.; Veglia, F.; Biunno, I.; Gavazzi, A.; Calabrese, E.; Martinelli Boneschi, F.; Sorbi, S.; Mariani, C.; Franceschi, M.; Grimaldi, L. M. Interleukin-1B polymorphism is associated with age at onset of Alzheimer's disease. Neurobiol Aging 2003, 24, 927-31. Scorziello, A.; Meucci, O.; Florio, T.; Fattore, M.; Forloni, G.; Salmona, M.; Schettini, G. beta 25-35 alters calcium homeostasis and induces neurotoxicity in cerebellar granule cells. J Neurochem 1996, 66, 1995-2003. Sczakiel, G. The design of antisense RNA. Antisense Nucleic Acid Drug Dev 1997, 7, 439-44. Seabrook, G. R.; Smith, D. W.; Bowery, B. J.; Easter, A.; Reynolds, T.; Fitzjohn, S. M.; Morton, R. A.; Zheng, H.; Dawson, G. R.; Sirinathsinghji, D. J.; Davies, C. H.; Collingridge, G. L.; Hill, R. G. Mechanisms contributing to the deficits in hippocampal synaptic plasticity in mice lacking amyloid precursor protein. Neuropharmacology 1999, 38, 349-59. Sehgal, P. B. Interleukin-6: molecular pathophysiology. J Invest Dermatol 1990, 94, 2S-6S. Sei, S.; Yang, Q. E.; O'Neill, D.; Yoshimura, K.; Nagashima, K.; Mitsuya, H. Identification of a key target sequence to block human immunodeficiency virus type 1 replication within the gag-pol transframe domain. J Virol 2000, 74, 4621-33. Selkoe, D. J. The molecular pathology of Alzheimer's disease. Neuron 1991, 6, 487-98. Selmaj, K. W.; Farooq, M.; Norton, W. T.; Raine, C. S.; Brosnan, C. F. Proliferation of astrocytes in vitro in response to cytokines. A primary role for tumor necrosis factor. J Immunol 1990, 144, 129-35. Sen, R.; Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 1986, 46, 705-16. Serafini, T.; Colamarino, S. A.; Leonardo, E. D.; Wang, H.; Beddington, R.; Skarnes, W. C.; TessierLavigne, M. Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system. Cell 1996, 87, 1001-14. Seubert, P.; Oltersdorf, T.; Lee, M. G.; Barbour, R.; Blomquist, C.; Davis, D. L.; Bryant, K.; Fritz, L. C.; Galasko, D.; Thal, L. J. Secretion of beta-amyloid precursor protein cleaved at the amino terminus of the beta-amyloid peptide. Nature 1993, 361, 260-3. Shaffer, L. M.; Dority, M. D.; Gupta-Bansal, R.; Frederickson, R. C.; Younkin, S. G.; Brunden, K. R. Amyloid beta protein (A beta) removal by neuroglial cells in culture. Neurobiol Aging 1995, 16, 737-45. Shearman, M. S.; Ragan, C. I.; Iversen, L. L. Inhibition of PC12 cell redox activity is a specific, early indicator of the mechanism of beta-amyloid-mediated cell death. Proc Natl Acad Sci U S A 1994, 91, 1470-4. Sheng, J. G.; Griffin, W. S.; Royston, M. C.; Mrak, R. E. Distribution of interleukin-1-immunoreactive microglia in cerebral cortical layers: implications for neuritic plaque formation in Alzheimer's disease. Neuropathol Appl Neurobiol 1998, 24, 278-83. Sheng, J. G.; Mrak, R. E.; Griffin, W. S. Glial-neuronal interactions in Alzheimer disease: progressive association of IL-1alpha+ microglia and S100beta+ astrocytes with neurofibrillary tangle stages. J Neuropathol Exp Neurol 1997, 56, 285-90. Sheppard, K. A.; Phelps, K. M.; Williams, A. J.; Thanos, D.; Glass, C. K.; Rosenfeld, M. G.; Gerritsen, M. E.; Collins, T. Nuclear integration of glucocorticoid receptor and nuclear factor-kappaB signaling by CREB-binding protein and steroid receptor coactivator-1. J Biol Chem 1998, 273, 29291-4. Sherrington, R.; Rogaev, E. I.; Liang, Y.; Rogaeva, E. A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; Holman, K. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature 1995, 375, 754-60. Shibasaki, F.; Price, E. R.; Milan, D.; McKeon, F. Role of kinases and the phosphatase calcineurin in the nuclear shuttling of transcription factor NF-AT4. Nature 1996, 382, 370-3. Shibata, N.; Ohnuma, T.; Takahashi, T.; Baba, H.; Ishizuka, T.; Ohtsuka, M.; Ueki, A.; Nagao, M.; Arai, H. Effect of IL-6 polymorphism on risk of Alzheimer disease: genotype-phenotype association study in Japanese cases. Am J Med Genet 2002, 114, 436-9. 80 REFERENCES L. Fisher Shie, F. S.; LeBoeur, R. C.; Jin, L. W. Early intraneuronal Abeta deposition in the hippocampus of APP transgenic mice. Neuroreport 2003, 14, 123-9. Shimizu, H.; Mitomo, K.; Watanabe, T.; Okamoto, S.; Yamamoto, K. Involvement of a NF-kappa B-like transcription factor in the activation of the interleukin-6 gene by inflammatory lymphokines. Mol Cell Biol 1990, 10, 561-8. Shimizu, H.; Yamamoto, K. NF-kappa B and C/EBP transcription factor families synergistically function in mouse serum amyloid A gene expression induced by inflammatory cytokines. Gene 1994, 149, 305-10. Shirakawa, F.; Saito, K.; Bonagura, C. A.; Galson, D. L.; Fenton, M. J.; Webb, A. C.; Auron, P. E. The human prointerleukin 1 beta gene requires DNA sequences both proximal and distal to the transcription start site for tissue-specific induction. Mol Cell Biol 1993, 13, 1332-44. Shors, T. J.; Miesegaes, G.; Beylin, A.; Zhao, M.; Rydel, T.; Gould, E. Neurogenesis in the adult is involved in the formation of trace memories. Nature 2001, 410, 372-6. Siebenlist, U.; Franzoso, G.; Brown, K. Structure, regulation and function of NF-kappa B. Annu Rev Cell Biol 1994, 10, 405-55. Sievers, J.; Parwaresch, R.; Wottge, H. U. Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes: morphology. Glia 1994, 12, 245-58. Silhol, M.; Tyagi, M.; Giacca, M.; Lebleu, B.; Vives, E. Different mechanisms for cellular internalization of the HIV-1 Tat-derived cell penetrating peptide and recombinant proteins fused to Tat. Eur J Biochem 2002, 269, 494-501. Silver, J.; Sapiro, J. Axonal guidance during development of the optic nerve: the role of pigmented epithelia and other extrinsic factors. J Comp Neurol 1981, 202, 521-38. Simic, G.; Lucassen, P. J.; Krsnik, Z.; Kruslin, B.; Kostovic, I.; Winblad, B.; Bogdanovi nNOS expression in reactive astrocytes correlates with increased cell death related DNA damage in the hippocampus and entorhinal cortex in Alzheimer's disease. Exp Neurol 2000, 165, 12-26. Sims, J. E.; Gayle, M. A.; Slack, J. L.; Alderson, M. R.; Bird, T. A.; Giri, J. G.; Colotta, F.; Re, F.; Mantovani, A.; Shanebeck, K.; et al. Interleukin 1 signaling occurs exclusively via the type I receptor. Proc Natl Acad Sci U S A 1993, 90, 6155-9. Sims, J. E.; March, C. J.; Cosman, D.; Widmer, M. B.; MacDonald, H. R.; McMahan, C. J.; Grubin, C. E.; Wignall, J. M.; Jackson, J. L.; Call, S. M.; et al. cDNA expression cloning of the IL-1 receptor, a member of the immunoglobulin superfamily. Science 1988, 241, 585-9. Singh, S.; Aggarwal, B. B. Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected]. J Biol Chem 1995a, 270, 24995-5000. Singh, S.; Aggarwal, B. B. Protein-tyrosine phosphatase inhibitors block tumor necrosis factor-dependent activation of the nuclear transcription factor NF-kappa B. J Biol Chem 1995b, 270, 10631-9. Sinha, S.; Anderson, J. P.; Barbour, R.; Basi, G. S.; Caccavello, R.; Davis, D.; Doan, M.; Dovey, H. F.; Frigon, N.; Hong, J.; Jacobson-Croak, K.; Jewett, N.; Keim, P.; Knops, J.; Lieberburg, I.; Power, M.; Tan, H.; Tatsuno, G.; Tung, J.; Schenk, D.; Seubert, P.; Suomensaari, S. M.; Wang, S.; Walker, D.; John, V. Purification and cloning of amyloid precursor protein beta-secretase from human brain. Nature 1999, 402, 537-40. Skaper, S. D.; Facci, L.; Leon, A. Inflammatory mediator stimulation of astrocytes and meningeal fibroblasts induces neuronal degeneration via the nitridergic pathway. J Neurochem 1995, 64, 26676. Small, D. H.; Mok, S. S.; Bornstein, J. C. Alzheimer's disease and Abeta toxicity: from top to bottom. Nat Rev Neurosci 2001, 2, 595-8. Smith, M. A. Alzheimer disease. Int Rev Neurobiol 1998, 42, 1-54. Smith, M. A.; Drew, K. L.; Nunomura, A.; Takeda, A.; Hirai, K.; Zhu, X.; Atwood, C. S.; Raina, A. K.; Rottkamp, C. A.; Sayre, L. M.; Friedland, R. P.; Perry, G. Amyloid-beta, tau alterations and mitochondrial dysfunction in Alzheimer disease: the chickens or the eggs? Neurochem Int 2002, 40, 527-31. Song, Y. S.; Park, H. J.; Kim, S. Y.; Lee, S. H.; Yoo, H. S.; Lee, H. S.; Lee, M. K.; Oh, K. W.; Kang, S. K.; Lee, S. E.; Hong, J. T. Protective role of Bcl-2 on beta-amyloid-induced cell death of differentiated PC12 cells: reduction of NF-kappaB and p38 MAP kinase activation. Neurosci Res 2004, 49, 69-80. REFERENCES 81 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Soomets, U.; Lindgren, M.; Gallet, X.; Hallbrink, M.; Elmquist, A.; Balaspiri, L.; Zorko, M.; Pooga, M.; Brasseur, R.; Langel, Ü. Deletion analogues of transportan. Biochim Biophys Acta 2000, 1467, 165-76. Sparacio, S. M.; Zhang, Y.; Vilcek, J.; Benveniste, E. N. Cytokine regulation of interleukin-6 gene expression in astrocytes involves activation of an NF-kappa B-like nuclear protein. J Neuroimmunol 1992, 39, 231-42. Srinivasan, S. K.; Iversen, P. Review of in vivo pharmacokinetics and toxicology of phosphorothioate oligonucleotides. J Clin Lab Anal 1995, 9, 129-37. St George-Hyslop, P. H. Genetic factors in the genesis of Alzheimer's disease. Ann N Y Acad Sci 2000, 924, 1-7. Stanley, E. R.; Berg, K. L.; Einstein, D. B.; Lee, P. S.; Pixley, F. J.; Wang, Y.; Yeung, Y. G. Biology and action of colony--stimulating factor-1. Mol Reprod Dev 1997, 46, 4-10. Stein, B.; Baldwin, A. S., Jr. Distinct mechanisms for regulation of the interleukin-8 gene involve synergism and cooperativity between C/EBP and NF-kappa B. Mol Cell Biol 1993, 13, 7191-8. Stein, B.; Baldwin, A. S., Jr.; Ballard, D. W.; Greene, W. C.; Angel, P.; Herrlich, P. Cross-coupling of the NF-kappa B p65 and Fos/Jun transcription factors produces potentiated biological function. Embo J 1993, 12, 3879-91. Stein, B.; Yang, M. X. Repression of the interleukin-6 promoter by estrogen receptor is mediated by NFkappa B and C/EBP beta. Mol Cell Biol 1995, 15, 4971-9. Stein, T. D.; Anders, N. J.; DeCarli, C.; Chan, S. L.; Mattson, M. P.; Johnson, J. A. Neutralization of transthyretin reverses the neuroprotective effects of secreted amyloid precursor protein (APP) in APPSW mice resulting in tau phosphorylation and loss of hippocampal neurons: support for the amyloid hypothesis. J Neurosci 2004, 24, 7707-17. Stewart, W. F.; Kawas, C.; Corrada, M.; Metter, E. J. Risk of Alzheimer's disease and duration of NSAID use. Neurology 1997, 48, 626-32. Streit, W. J.; Walter, S. A.; Pennell, N. A. Reactive microgliosis. Prog Neurobiol 1999, 57, 563-81. Subbarao, K. V.; Richardson, J. S.; Ang, L. C. Autopsy samples of Alzheimer's cortex show increased peroxidation in vitro. J Neurochem 1990, 55, 342-5. Sui, G.; Soohoo, C.; Affar el, B.; Gay, F.; Shi, Y.; Forrester, W. C. A DNA vector-based RNAi technology to suppress gene expression in mammalian cells. Proc Natl Acad Sci U S A 2002, 99, 5515-20. Sunde, M.; Blake, C. C. From the globular to the fibrous state: protein structure and structural conversion in amyloid formation. Q Rev Biophys 1998, 31, 1-39. Suzuki, N.; Cheung, T. T.; Cai, X. D.; Odaka, A.; Otvos, L., Jr.; Eckman, C.; Golde, T. E.; Younkin, S. G. An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science 1994, 264, 1336-40. Suzuki, T.; Futaki, S.; Niwa, M.; Tanaka, S.; Ueda, K.; Sugiura, Y. Possible existence of common internalization mechanisms among arginine-rich peptides. J Biol Chem 2002, 277, 2437-43. Suzuki, T.; Wu, D.; Schlachetzki, F.; Li, J. Y.; Boado, R. J.; Pardridge, W. M. Imaging Endogenous Gene Expression in Brain Cancer In Vivo with 111In-Peptide Nucleic Acid Antisense Radiopharmaceuticals and Brain Drug-Targeting Technology. J Nucl Med 2004, 45, 1766-75. Taga, T.; Kishimoto, T. Gp130 and the interleukin-6 family of cytokines. Annu Rev Immunol 1997, 15, 797-819. Takada, Y.; Bhardwaj, A.; Potdar, P.; Aggarwal, B. B. Nonsteroidal anti-inflammatory agents differ in their ability to suppress NF-kappaB activation, inhibition of expression of cyclooxygenase-2 and cyclin D1, and abrogation of tumor cell proliferation. Oncogene 2004a, 23, 9247-58. Takada, Y.; Singh, S.; Aggarwal, B. B. Identification of a p65 peptide that selectively inhibits NF-kappa B activation induced by various inflammatory stimuli and its role in down-regulation of NF-kappaBmediated gene expression and up-regulation of apoptosis. J Biol Chem 2004b, 279, 15096-104. Tanaka, M.; Fuentes, M. E.; Yamaguchi, K.; Durnin, M. H.; Dalrymple, S. A.; Hardy, K. L.; Goeddel, D. V. Embryonic lethality, liver degeneration, and impaired NF-kappa B activation in IKK-betadeficient mice. Immunity 1999, 10, 421-9. Tanzi, R. E.; Gusella, J. F.; Watkins, P. C.; Bruns, G. A.; St George-Hyslop, P.; Van Keuren, M. L.; Patterson, D.; Pagan, S.; Kurnit, D. M.; Neve, R. L. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science 1987, 235, 880-4. 82 REFERENCES L. Fisher Tanzi, R. E.; McClatchey, A. I.; Lamperti, E. D.; Villa-Komaroff, L.; Gusella, J. F.; Neve, R. L. Protease inhibitor domain encoded by an amyloid protein precursor mRNA associated with Alzheimer's disease. Nature 1988, 331, 528-30. Tarkowski, E.; Liljeroth, A. M.; Minthon, L.; Tarkowski, A.; Wallin, A.; Blennow, K. Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res Bull 2003, 61, 255-60. Tchelingerian, J. L.; Le Saux, F.; Jacque, C. Identification and topography of neuronal cell populations expressing TNF alpha and IL-1 alpha in response to hippocampal lesion. J Neurosci Res 1996, 43, 99-106. Tehranian, R.; Hasanvan, H.; Iverfeldt, K.; Post, C.; Schultzberg, M. Early induction of interleukin-6 mRNA in the hippocampus and cortex of APPsw transgenic mice Tg2576. Neurosci Lett 2001, 301, 54-8. Ten, R. M.; Paya, C. V.; Israel, N.; Le Bail, O.; Mattei, M. G.; Virelizier, J. L.; Kourilsky, P.; Israel, A. The characterization of the promoter of the gene encoding the p50 subunit of NF-kappa B indicates that it participates in its own regulation. Embo J 1992, 11, 195-203. Terai, K.; Matsuo, A.; McGeer, P. L. Enhancement of immunoreactivity for NF-kappa B in the hippocampal formation and cerebral cortex of Alzheimer's disease. Brain Res 1996, 735, 159-68. Terzi, E.; Holzemann, G.; Seelig, J. Alzheimer beta-amyloid peptide 25-35: electrostatic interactions with phospholipid membranes. Biochemistry 1994, 33, 7434-41. Tessier, D. J.; Komalavilas, P.; Liu, B.; Kent, C. K.; Thresher, J. S.; Dreiza, C. M.; Panitch, A.; Joshi, L.; Furnish, E.; Stone, W.; Fowl, R.; Brophy, C. M. Transduction of peptide analogs of the small heat shock-related protein HSP20 inhibits intimal hyperplasia. J Vasc Surg 2004, 40, 106-14. Thal, D. R.; Schultz, C.; Dehghani, F.; Yamaguchi, H.; Braak, H.; Braak, E. Amyloid beta-protein (Abeta)containing astrocytes are located preferentially near N-terminal-truncated Abeta deposits in the human entorhinal cortex. Acta Neuropathol (Berl) 2000, 100, 608-17. Thangnipon, W.; Kingsbury, A.; Webb, M.; Balazs, R. Observations on rat cerebellar cells in vitro: influence of substratum, potassium concentration and relationship between neurones and astrocytes. Brain Res 1983, 313, 177-89. Thanos, D.; Maniatis, T. Virus induction of human IFN beta gene expression requires the assembly of an enhanceosome. Cell 1995, 83, 1091-100. Tomaselli, K. J.; Neugebauer, K. M.; Bixby, J. L.; Lilien, J.; Reichardt, L. F. N-cadherin and integrins: two receptor systems that mediate neuronal process outgrowth on astrocyte surfaces. Neuron 1988, 1, 33-43. Tomidokoro, Y.; Ishiguro, K.; Harigaya, Y.; Matsubara, E.; Ikeda, M.; Park, J. M.; Yasutake, K.; Kawarabayashi, T.; Okamoto, K.; Shoji, M. Abeta amyloidosis induces the initial stage of tau accumulation in APP(Sw) mice. Neurosci Lett 2001, 299, 169-72. Tomita, N.; Morishita, R.; Tomita, S.; Gibbons, G. H.; Zhang, L.; Horiuchi, M.; Kaneda, Y.; Higaki, J.; Ogihara, T.; Dzau, V. J. Transcription factor decoy for NFkappaB inhibits TNF-alpha-induced cytokine and adhesion molecule expression in vivo. Gene Ther 2000a, 7, 1326-32. Tomita, N.; Morishita, R.; Tomita, S.; Yamamoto, K.; Aoki, M.; Matsushita, H.; Hayashi, S.; Higaki, J.; Ogihara, T. Transcription factor decoy for nuclear factor-kappaB inhibits tumor necrosis factoralpha-induced expression of interleukin-6 and intracellular adhesion molecule-1 in endothelial cells. J Hypertens 1998, 16, 993-1000. Tomita, S.; Fujita, T.; Kirino, Y.; Suzuki, T. PDZ domain-dependent suppression of NF-kappaB/p65induced Abeta42 production by a neuron-specific X11-like protein. J Biol Chem 2000b, 275, 13056-60. Tomita, T.; Takano, H.; Tomita, N.; Morishita, R.; Kaneko, M.; Shi, K.; Takahi, K.; Nakase, T.; Kaneda, Y.; Yoshikawa, H.; Ochi, T. Transcription factor decoy for NFkappaB inhibits cytokine and adhesion molecule expressions in synovial cells derived from rheumatoid arthritis. Rheumatology (Oxford) 2000c, 39, 749-57. Toro, V. C.; Tehranian, R.; Zetterstrom, M.; Eriksson, G.; Langel, U.; Bartfai, T.; Iverfeld, K. Increased gene expression of interleukin-1alpha and interleukin-6 in rat primary glial cells induced by betaamyloid fragment. J Mol Neurosci 2001, 17, 341-50. Touzani, O.; Boutin, H.; Chuquet, J.; Rothwell, N. Potential mechanisms of interleukin-1 involvement in cerebral ischaemia. J Neuroimmunol 1999, 100, 203-15. REFERENCES 83 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Turner, R. S.; Suzuki, N.; Chyung, A. S.; Younkin, S. G.; Lee, V. M. Amyloids beta40 and beta42 are generated intracellularly in cultured human neurons and their secretion increases with maturation. J Biol Chem 1996, 271, 8966-70. Tyler, B. M.; Jansen, K.; McCormick, D. J.; Douglas, C. L.; Boules, M.; Stewart, J. A.; Zhao, L.; Lacy, B.; Cusack, B.; Fauq, A.; Richelson, E. Peptide nucleic acids targeted to the neurotensin receptor and administered i.p. cross the blood-brain barrier and specifically reduce gene expression. Proc Natl Acad Sci U S A 1999, 96, 7053-8. Tyler, B. M.; McCormick, D. J.; Hoshall, C. V.; Douglas, C. L.; Jansen, K.; Lacy, B. W.; Cusack, B.; Richelson, E. Specific gene blockade shows that peptide nucleic acids readily enter neuronal cells in vivo. FEBS Lett 1998, 421, 280-4. Tyler-McMahon, B. M.; Stewart, J. A.; Jackson, J.; Bitner, M. D.; Fauq, A.; McCormick, D. J.; Richelson, E. Altering behavioral responses and dopamine transporter protein with antisense peptide nucleic acids. Biochem Pharmacol 2001, 62, 929-32. Vallieres, L.; Campbell, I. L.; Gage, F. H.; Sawchenko, P. E. Reduced hippocampal neurogenesis in adult transgenic mice with chronic astrocytic production of interleukin-6. J Neurosci 2002, 22, 486-92. Van Antwerp, D. J.; Martin, S. J.; Kafri, T.; Green, D. R.; Verma, I. M. Suppression of TNF-alpha-induced apoptosis by NF-kappaB. Science 1996, 274, 787-9. Van Wagoner, N. J.; Oh, J. W.; Repovic, P.; Benveniste, E. N. Interleukin-6 (IL-6) production by astrocytes: autocrine regulation by IL-6 and the soluble IL-6 receptor. J Neurosci 1999, 19, 523644. Vanden Berghe, W.; De Bosscher, K.; Boone, E.; Plaisance, S.; Haegeman, G. The nuclear factor-kappaB engages CBP/p300 and histone acetyltransferase activity for transcriptional activation of the interleukin-6 gene promoter. J Biol Chem 1999, 274, 32091-8. Vandenabeele, P.; Fiers, W. Is amyloidogenesis during Alzheimer's disease due to an IL-1-/IL-6-mediated 'acute phase response' in the brain? Immunol Today 1991, 12, 217-9. Varadarajan, S.; Kanski, J.; Aksenova, M.; Lauderback, C.; Butterfield, D. A. Different mechanisms of oxidative stress and neurotoxicity for Alzheimer's A beta(1--42) and A beta(25--35). J Am Chem Soc 2001, 123, 5625-31. Varadarajan, S.; Yatin, S.; Kanski, J.; Jahanshahi, F.; Butterfield, D. A. Methionine residue 35 is important in amyloid beta-peptide-associated free radical oxidative stress. Brain Res Bull 1999, 50, 133-41. Vassar, R.; Bennett, B. D.; Babu-Khan, S.; Kahn, S.; Mendiaz, E. A.; Denis, P.; Teplow, D. B.; Ross, S.; Amarante, P.; Loeloff, R.; Luo, Y.; Fisher, S.; Fuller, J.; Edenson, S.; Lile, J.; Jarosinski, M. A.; Biere, A. L.; Curran, E.; Burgess, T.; Louis, J. C.; Collins, F.; Treanor, J.; Rogers, G.; Citron, M. Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science 1999, 286, 735-41. Vehmas, A. K.; Kawas, C. H.; Stewart, W. F.; Troncoso, J. C. Immune reactive cells in senile plaques and cognitive decline in Alzheimer's disease. Neurobiol Aging 2003, 24, 321-31. Vickers, T. A.; Griffith, M. C.; Ramasamy, K.; Risen, L. M.; Freier, S. M. Inhibition of NF-kappa B specific transcriptional activation by PNA strand invasion. Nucleic Acids Res 1995, 23, 3003-8. von Bernhardi, R.; Eugenin, J. Microglial reactivity to beta-amyloid is modulated by astrocytes and proinflammatory factors. Brain Res 2004, 1025, 186-93. von Zahn, J.; Moller, T.; Kettenmann, H.; Nolte, C. Microglial phagocytosis is modulated by pro- and antiinflammatory cytokines. Neuroreport 1997, 8, 3851-6. Walker, D. G.; Kim, S. U.; McGeer, P. L. Expression of complement C4 and C9 genes by human astrocytes. Brain Res 1998, 809, 31-8. Walsh, D. M.; Hartley, D. M.; Kusumoto, Y.; Fezoui, Y.; Condron, M. M.; Lomakin, A.; Benedek, G. B.; Selkoe, D. J.; Teplow, D. B. Amyloid beta-protein fibrillogenesis. Structure and biological activity of protofibrillar intermediates. J Biol Chem 1999, 274, 25945-52. Walsh, D. M.; Klyubin, I.; Fadeeva, J. V.; Rowan, M. J.; Selkoe, D. J. Amyloid-beta oligomers: their production, toxicity and therapeutic inhibition. Biochem Soc Trans 2002, 30, 552-7. Walsh, D. M.; Lomakin, A.; Benedek, G. B.; Condron, M. M.; Teplow, D. B. Amyloid beta-protein fibrillogenesis. Detection of a protofibrillar intermediate. J Biol Chem 1997, 272, 22364-72. Wang, C. Y.; Mayo, M. W.; Baldwin, A. S., Jr. TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB. Science 1996a, 274, 784-7. 84 REFERENCES L. Fisher Wang, H. Y.; Lee, D. H.; Davis, C. B.; Shank, R. P. Amyloid peptide Abeta(1-42) binds selectively and with picomolar affinity to alpha7 nicotinic acetylcholine receptors. J Neurochem 2000, 75, 115561. Wang, R.; Sweeney, D.; Gandy, S. E.; Sisodia, S. S. The profile of soluble amyloid beta protein in cultured cell media. Detection and quantification of amyloid beta protein and variants by immunoprecipitation-mass spectrometry. J Biol Chem 1996b, 271, 31894-902. Weaver, J. D.; Huang, M. H.; Albert, M.; Harris, T.; Rowe, J. W.; Seeman, T. E. Interleukin-6 and risk of cognitive decline: MacArthur studies of successful aging. Neurology 2002, 59, 371-8. Wegiel, J.; Imaki, H.; Wang, K. C.; Wronska, A.; Osuchowski, M.; Rubenstein, R. Origin and turnover of microglial cells in fibrillar plaques of APPsw transgenic mice. Acta Neuropathol (Berl) 2003, 105, 393-402. Wegiel, J.; Wang, K. C.; Imaki, H.; Rubenstein, R.; Wronska, A.; Osuchowski, M.; Lipinski, W. J.; Walker, L. C.; LeVine, H. The role of microglial cells and astrocytes in fibrillar plaque evolution in transgenic APP(SW) mice. Neurobiol Aging 2001, 22, 49-61. Weldon, D. T.; Rogers, S. D.; Ghilardi, J. R.; Finke, M. P.; Cleary, J. P.; O'Hare, E.; Esler, W. P.; Maggio, J. E.; Mantyh, P. W. Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J Neurosci 1998, 18, 2161-73. Wender, P. A.; Mitchell, D. J.; Pattabiraman, K.; Pelkey, E. T.; Steinman, L.; Rothbard, J. B. The design, synthesis, and evaluation of molecules that enable or enhance cellular uptake: peptoid molecular transporters. Proc Natl Acad Sci U S A 2000, 97, 13003-8. Wertkin, A. M.; Turner, R. S.; Pleasure, S. J.; Golde, T. E.; Younkin, S. G.; Trojanowski, J. Q.; Lee, V. M. Human neurons derived from a teratocarcinoma cell line express solely the 695-amino acid amyloid precursor protein and produce intracellular beta-amyloid or A4 peptides. Proc Natl Acad Sci U S A 1993, 90, 9513-7. Wilcox, C. M.; Alexander, L. N.; Cotsonis, G. A.; Clark, W. S. Nonsteroidal antiinflammatory drugs are associated with both upper and lower gastrointestinal bleeding. Dig Dis Sci 1997, 42, 990-7. Wilson, C. A.; Doms, R. W.; Zheng, H.; Lee, V. M. Presenilins are not required for A beta 42 production in the early secretory pathway. Nat Neurosci 2002a, 5, 849-55. Wilson, C. J.; Finch, C. E.; Cohen, H. J. Cytokines and cognition--the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc 2002b, 50, 2041-56. Wirths, O.; Multhaup, G.; Czech, C.; Blanchard, V.; Moussaoui, S.; Tremp, G.; Pradier, L.; Beyreuther, K.; Bayer, T. A. Intraneuronal Abeta accumulation precedes plaque formation in beta-amyloid precursor protein and presenilin-1 double-transgenic mice. Neurosci Lett 2001, 306, 116-20. Wischik, C. M.; Novak, M.; Thogersen, H. C.; Edwards, P. C.; Runswick, M. J.; Jakes, R.; Walker, J. E.; Milstein, C.; Roth, M.; Klug, A. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease. Proc Natl Acad Sci U S A 1988, 85, 4506-10. Wisniewski, T.; Ghiso, J.; Frangione, B. Peptides homologous to the amyloid protein of Alzheimer's disease containing a glutamine for glutamic acid substitution have accelerated amyloid fibril formation. Biochem Biophys Res Commun 1991, 179, 1247-54. Wittung, P.; Kajanus, J.; Edwards, K.; Haaima, G.; Nielsen, P. E.; Norden, B.; Malmstrom, B. G. Phospholipid membrane permeability of peptide nucleic acid. FEBS Lett 1995, 375, 27-9. Wyss-Coray, T.; Loike, J. D.; Brionne, T. C.; Lu, E.; Anankov, R.; Yan, F.; Silverstein, S. C.; Husemann, J. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med 2003, 9, 453-7. Wyss-Coray, T.; McConlogue, L.; Kindy, M.; Schmidt, A. M.; Du Yan, S.; Stern, D. M. Key signaling pathways regulate the biological activities and accumulation of amyloid-beta. Neurobiol Aging 2001, 22, 967-73. Xia, C.; Cheshire, J. K.; Patel, H.; Woo, P. Cross-talk between transcription factors NF-kappa B and C/EBP in the transcriptional regulation of genes. Int J Biochem Cell Biol 1997, 29, 1525-39. Xie, Z.; Wei, M.; Morgan, T. E.; Fabrizio, P.; Han, D.; Finch, C. E.; Longo, V. D. Peroxynitrite mediates neurotoxicity of amyloid beta-peptide1-42- and lipopolysaccharide-activated microglia. J Neurosci 2002, 22, 3484-92. Yaar, M.; Zhai, S.; Pilch, P. F.; Doyle, S. M.; Eisenhauer, P. B.; Fine, R. E.; Gilchrest, B. A. Binding of beta-amyloid to the p75 neurotrophin receptor induces apoptosis. A possible mechanism for Alzheimer's disease. J Clin Invest 1997, 100, 2333-40. REFERENCES 85 Inflammatory Cytokines and NFκB in Alzheimer’s Disease Yamaguchi, H.; Sugihara, S.; Ogawa, A.; Saido, T. C.; Ihara, Y. Diffuse plaques associated with astroglial amyloid beta protein, possibly showing a disappearing stage of senile plaques. Acta Neuropathol (Berl) 1998, 95, 217-22. Yamamoto, Y.; Yin, M. J.; Lin, K. M.; Gaynor, R. B. Sulindac inhibits activation of the NF-kappaB pathway. J Biol Chem 1999, 274, 27307-14. Yan, R.; Bienkowski, M. J.; Shuck, M. E.; Miao, H.; Tory, M. C.; Pauley, A. M.; Brashier, J. R.; Stratman, N. C.; Mathews, W. R.; Buhl, A. E.; Carter, D. B.; Tomasselli, A. G.; Parodi, L. A.; Heinrikson, R. L.; Gurney, M. E. Membrane-anchored aspartyl protease with Alzheimer's disease betasecretase activity. Nature 1999a, 402, 533-7. Yan, S. D.; Chen, X.; Fu, J.; Chen, M.; Zhu, H.; Roher, A.; Slattery, T.; Zhao, L.; Nagashima, M.; Morser, J.; Migheli, A.; Nawroth, P.; Stern, D.; Schmidt, A. M. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. Nature 1996, 382, 685-91. Yan, S. D.; Chen, X.; Schmidt, A. M.; Brett, J.; Godman, G.; Zou, Y. S.; Scott, C. W.; Caputo, C.; Frappier, T.; Smith, M. A.; et al. Glycated tau protein in Alzheimer disease: a mechanism for induction of oxidant stress. Proc Natl Acad Sci U S A 1994, 91, 7787-91. Yan, S. D.; Roher, A.; Schmidt, A. M.; Stern, D. M. Cellular cofactors for amyloid beta-peptide-induced cell stress. Moving from cell culture to in vivo. Am J Pathol 1999b, 155, 1403-11. Yankner, B. A. Mechanisms of neuronal degeneration in Alzheimer's disease. Neuron 1996, 16, 921-32. Yasojima, K.; Schwab, C.; McGeer, E. G.; McGeer, P. L. Up-regulated production and activation of the complement system in Alzheimer's disease brain. Am J Pathol 1999, 154, 927-36. Yazawa, H.; Yu, Z. X.; Takeda; Le, Y.; Gong, W.; Ferrans, V. J.; Oppenheim, J. J.; Li, C. C.; Wang, J. M. Beta amyloid peptide (Abeta42) is internalized via the G-protein-coupled receptor FPRL1 and forms fibrillar aggregates in macrophages. Faseb J 2001, 15, 2454-62. Ye, S. M.; Johnson, R. W. Increased interleukin-6 expression by microglia from brain of aged mice. J Neuroimmunol 1999, 93, 139-48. Ye, S. M.; Johnson, R. W. Regulation of interleukin-6 gene expression in brain of aged mice by nuclear factor kappaB. J Neuroimmunol 2001, 117, 87-96. Yin, M. J.; Yamamoto, Y.; Gaynor, R. B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. Nature 1998, 396, 77-80. Yu, C.; Kim, S.H.; Ikeuchi, T.; Xu, H.; Gasparini, L.; Wang, R.; Sisodia, S.S. Characterization of a presenilin-mediated amyloid precursor protein carboxyl-terminal fragment γ. Evidence for distinct mechanisms involved in γ-secretase processing of the APP and Notch1 transmembrane domains. J Biol Chem 2001, 276, 43756-60. Yu, W. H.; Go, L.; Guinn, B. A.; Fraser, P. E.; Westaway, D.; McLaurin, J. Phenotypic and functional changes in glial cells as a function of age. Neurobiol Aging 2002, 23, 105-15. Zhang, C.; Lambert, M. P.; Bunch, C.; Barber, K.; Wade, W. S.; Krafft, G. A.; Klein, W. L. Focal adhesion kinase expressed by nerve cell lines shows increased tyrosine phosphorylation in response to Alzheimer's A beta peptide. J Biol Chem 1994, 269, 25247-50. Zhang, Y.; McLaughlin, R.; Goodyer, C.; LeBlanc, A. Selective cytotoxicity of intracellular amyloid beta peptide1-42 through p53 and Bax in cultured primary human neurons. J Cell Biol 2002, 156, 51929. Zhang, Z.; Nadeau, P.; Song, W.; Donoviel, D.; Yuan, M.; Bernstein, A.; Yankner, B. A. Presenilins are required for gamma-secretase cleavage of beta-APP and transmembrane cleavage of Notch-1. Nat Cell Biol 2000, 2, 463-5. Zhao, G.; Mao, G.; Tan, J.; Dong, Y.; Cui, M.Z.; Kim, S.H.; Xu, X. Identification of a new presenilindependent zeta-cleavage site within the transmembrane domain of amyloid precursor protein. J Biol Chem 2004, 49, 50647-50. Zheng, H.; Jiang, M.; Trumbauer, M. E.; Sirinathsinghji, D. J.; Hopkins, R.; Smith, D. W.; Heavens, R. P.; Dawson, G. R.; Boyce, S.; Conner, M. W. beta-Amyloid precursor protein-deficient mice show reactive gliosis and decreased locomotor activity. Cell 1995, 81, 525-31. Zirrgiebel, U.; Ohga, Y.; Carter, B.; Berninger, B.; Inagaki, N.; Thoenen, H.; Lindholm, D. Characterization of TrkB receptor-mediated signaling pathways in rat cerebellar granule neurons: involvement of protein kinase C in neuronal survival. J Neurochem 1995, 65, 2241-50. Östenson, C. G.; Sandberg-Nordqvist, A. C.; Chen, J.; Hallbrink, M.; Rotin, D.; Langel, Ü.; Efendic, S. Overexpression of protein-tyrosine phosphatase PTP sigma is linked to impaired glucose-induced 86 REFERENCES L. Fisher insulin secretion in hereditary diabetic Goto-Kakizaki rats. Biochem Biophys Res Commun 2002, 291, 945-50. REFERENCES 87
© Copyright 2025 Paperzz