THE JOURNAL OF BIOLOGICAL CHEMISTRY © 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Vol. 280, No. 17, Issue of April 29, pp. 17084 –17092, 2005 Printed in U.S.A. Small Interference RNA-mediated Gene Silencing of Human Biliverdin Reductase, but Not That of Heme Oxygenase-1, Attenuates Arsenite-mediated Induction of the Oxygenase and Increases Apoptosis in 293A Kidney Cells* Received for publication, November 19, 2004, and in revised form, February 28, 2005 Published, JBC Papers in Press, February 28, 2005, DOI 10.1074/jbc.M413121200 Tihomir Miralem, Zhenbo Hu, Michael D. Torno, Katherine M. Lelli, and Mahin D. Maines‡ From the Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York 14624 BVR reduces biliverdin, the HO-1 and HO-2 product, to bilirubin. Human biliverdin (BVR) is a serine/threonine kinase activated by free radicals. It is a leucine zipper (bZip) DNA-binding protein and a regulatory factor for 8/7-bp AP-1-regulated genes, including HO-1 and ATF-2/CREB. Presently, small interference (si) RNA constructs were used to investigate the role of human BVR in sodium arsenite (As)-mediated induction of HO-1 and in cytoprotection against apoptosis. Activation of BVR involved increased serine/threonine phosphorylation but not its protein or transcript levels. The peak activity at 1 h (4 –5-fold) after treatment of 293A cells with 5 M As preceded induction of HO-1 expression by 3 h. The following suggests BVR involvement in regulating oxidative stress response of HO-1: siBVR attenuated Asmediated increase in HO-1 expression; siBVR, but not siHO-1, inhibited As-dependent increased c-jun promoter activity; treatment of cells with As increased AP-1 binding of nuclear proteins; BVR was identified in the DNA-protein complex; and AP-1 binding of the in vitro translated BVR was phosphorylation-dependent and was attenuated by biliverdin. Most unexpectedly, cells transfected with siBVR, but not siHO-1, displayed a 4-fold increase in apoptotic cells when treated with 10 M As as detected by flow cytometry. The presence of BVR small interference RNA augmented the effect of As on levels of cytochrome c, TRAIL, and DR-5 mRNA and cleavage of poly(ADP-ribose) polymerase. The findings describe the function of BVR in HO-1 oxidative response and, demonstrate, for the first time, not only that BVR advances the role of HO-1 in cytoprotection but also affords cytoprotection independent of heme degradation. The final step in heme metabolism is carried out by the soluble enzyme biliverdin IX␣ reductase (BVR)1 (1, 2). A second * This work was supported by NIEHS Grants RO1/ES04066 and RO1/ES12187 from the National Institutes of Health. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. ‡ To whom correspondence should be addressed: Dept. of Biochemistry and Biophysics, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14624. Tel.: 585-275-5383; Fax: 585-2756007; E-mail: [email protected]. 1 The abbreviations used are: BVR, biliverdin IX␣ reductase; As, sodium arsenite; ATF-2, activating transcription factor-2; hBVR, human biliverdin IX␣ reductase; AP-1, activator protein-1; bZip, leucine zipper; CRE, cAMP-response element; CREB, cAMP-response elementbinding protein; heme, Fe-protoporphyrin IX; DR-5, death receptor-5; HO, heme oxygenase; siRNA, small interference RNA; ELISA, enzyme- form, biliverdin IX reductase, which is identical to NADPHflavin reductase, has also been described (3– 6); this form is active only during fetal development and has little, if any, similarity to BVR. BVR, an evolutionarily conserved enzyme with unique dual pH/cofactor-dependent activity profile, is primarily found in the mammalian species and is responsible for the production of bilirubin IX␣. In most other animal species, biliverdin constitutes the end product of heme (Fe-protoporphyrin) macro-cycle cleavage. BVR catalyzes the reduction of biliverdin by incorporating two hydrogen atoms derived from pyridine nucleotides (NADH in acidic pH or NADPH in basic pH range) into the open tetrapyrrole ring structure (1, 2). Biliverdin IX␣ in turn is a product of the mixed function oxidase activity of two forms of heme oxygenase proteins, HO-1 and HO-2 (7); the HO enzymes are also known as the HSP32 family of stress/heat shock proteins (8 –10). HO-1 is the stressresponsive cognate and is induced by a wide array of stimuli that have in common the ability to cause oxidative stress (reviewed in Refs. 7 and 11). Whereas in the past BVR was solely considered in the context of disposal of biliverdin, and bilirubin was considered a toxic waste product of the heme metabolic pathway, recent findings have alluded to notable biological functions of heme degradation products in the cell; bilirubin is an effective intracellular antioxidant and a modulator of signal transduction pathways and immune effector functions (12–16). Bilirubin and biliverdin inhibit phosphotransferase activity of protein kinase C and cAMP-dependent protein kinase (17); bilirubin inhibits BVR activity, and biliverdin inhibits HO activity (1, 18). Because of the noted activities of bile pigments, induction of HO-1 is now generally accepted to be a key component of anti-inflammatory/anti-apoptotic stress effector pathways. In addition, the recently ascribed functions to CO (19 –21), which result from oxidation of ␣-meso carbon bridge of heme when the tetrapyrrole is cleaved by HO enzymes, support cytoprotective functions of heme oxidation products. BVR, however, has functions distinct from its reductase activity and separate from those of its substrate and product; the protein is a serine/threonine kinase that is activated in response to hydroxyl radicals (22). BVR is a member of the “leucine zipper” (bZip) family of mammalian and yeast transcription factors that include c-Fos, c-Jun, ATF-2/CREB, Bach 1, P45, NrF2 and -3, (bZip), sYAP, GCN4, and c-Myc (23–32); the reductase in a homodimeric form binds to the 7- and 8-bp AP-1 and cAMP target genes, among them being ATF-2/CREB linked immunosorbent assay; EGFP, enhanced green fluorescent protein; oligo, oligonucleotide; PBS, phosphate-buffered saline; Pipes, 1,4piperazinediethanesulfonic acid; PARP, poly(ADP-ribose) polymerase; wt, wild type. 17084 This paper is available on line at http://www.jbc.org siRNA-mediated Gene Silencing of Biliverdin Reductase and c-jun (33). Arsenite is a reported regulator of bZip factors (44). BVR being a bZip transcription factor and activator of HO-1 could be among the list genes that are modulated by arsenite treatment. This is consistent with the observation that the reductase translocates from cytosol into the nucleus in response to HO-1 inducers, including free radicals of cyclic hydrocarbons, cytokines, and cGMP (34). The induction of HO-1 by the free radical generator, menadione, is attenuated in the presence of antisense BVR (30). Arsenite modulates the profile of HO-1 gene expression in the cell by activating a number of bZip transcription factors involved in stress signaling pathways including the mitogenactivated protein family of kinases, p70 (s6k), protein kinase C, and NF- as well as changes in cellular free Ca2⫹ and NO䡠 levels/production (35– 40). HO-1 and genes that are involved in apoptosis are among the targets for arsenite (9, 41– 45). Arsenite activation and increased binding of a complex of proteins, which includes the Jun protein, to the CRE/AP-1 site of the HO-1 promoter are essential for stress response of the oxygenase to this stimulus as well as to other oxidants. Furthermore, arsenic compounds are considered to be tumorigenic, and evidence points to their activity to disrupt normal control of apoptosis by inhibiting c-Jun N-terminal kinase phosphatase and activating the NF- signaling pathway (35, 39, 46). The effect arsenite has on cell death, however, is concentration-dependent and has been proposed to involve oxygen free radicals (47). The features of BVR that communicate with other proteins known to have gene regulatory function in the cell together with above noted observations support the potential role of BVR in the stress response of HO-1. The present study was undertaken to define further the role of BVR in the response of HO-1 to arsenite and to examine whether BVR itself has input into the cellular anti-apoptotic effector pathways. We show, using arsenite as the stimulus, that BVR is a key component of HO-1 stress response and that this involves activity of the reductase as a kinase, transcription factor, and the inactivator of biliverdin. We further demonstrate for the first time that BVR offers cytoprotection against cell death independent of HO-1 expression. MATERIALS AND METHODS Constructs—siBVR and siHO-1 motifs were selected according to the amino acid Asn-19 rule by finding the pattern in human BVR and human HO-1 cDNA sequences (48, 49). The target sequence for hBVR is located 96 bp downstream of the start codon (nucleotides 96 –116). For HO-1, the targeting sequence is 106 –126 nucleotides downstream of the start codon. A retroviral based vector pSuper-Retro for siRNA expression was purchased from OligoEngine Co. (Seattle, WA). Retrovirus/siRNA-expressing vectors pSuper-Retro-siBVR and pSuper-Retro-siHO-1 were constructed according to the manufacturer’s instructions. Briefly, oligos containing the sequence of 21-mer small interference RNA were synthesized as follows: BVR siRNA, 5⬘-GATCCCC (TCC TCA GCG TTC CTG AAC CTG) TTCAAGAGA (CAG GTT CAG GAA CGC TGA GGA) TTTTTGGAAA and 3⬘-GGG (AGG AGT CGC AAG GAC TTG GAC) AAGTTCTCT (GTC CAA GTC CTT GCG ACT CCT)AAAAACCTTTTCGA; HO-1 siRNA, 5⬘-GATCCCC (AAC TTT CAG AAG GGC CAG GTG) TTCAAGAGA (CAC CTG GCC CTT CTG AAA GTT) TTTTTGGAAA and 3⬘-GGG (TTG AAA GTC TTC CCG GTC CAC) AAGTTCTCT (GTG GAC CGG GAA GAC TTT CAA)AAAAACCTTTTCGA. Complementary oligos were gel-purified and annealed to form double-stranded DNA. To insert the oligos into the pSuper-Retro plasmid, BglII and HindIII were used to digest the vector, and the digested vector was gel-purified. The double-stranded oligos were phosphorylated by T4 kinase. Ligation of the vector and oligos was carried out by incubation of 10 l of reaction containing 2 l of vector, 1 l of oligo, 2 l of 5⫻ ligation buffer, and 1 l (1 unit) of T4 ligation buffer for 1 h at room temperature. 2 l of the ligation reaction was then used to transform DH5␣ Escherichia coli. Clones containing the oligo inserts were identified by restriction digest analysis. Selected positive clones were further verified by DNA sequencing. The verified constructs were named pSuper-Retro-siBVR and pSuper-Retro-siHO-1. The retroviral 17085 vectors generating BVR siRNA and HO-1 siRNA were transfected into the 293A packaging cell line, and the supernatant containing the expressed siBVR or siHO-1 retrovirus was then titrated by using NIH3T3 cell line. The concentration of retrovirus expressing siRNA used to infect cells in further experiments was at a multiplicity of infection of 4 plaque-forming units/cell. Other constructs used in this study are as follows. The expression vectors pEGFP-FLAG-hBVR and pEGFP-FLAG-hHO-1 were generated by inserting full-length human BVR cDNA or full-length human HO-1 cDNA, respectively, into plasmid pEGFP (Clontech). The expression vectors generate an EGFP-FLAG-hBVR fusion protein or an EGFPFLAG-hHO-1 fusion protein. The vectors used for luciferase reporter assay are as follows: a pGL3 luciferase-containing vector ligated with a c-jun promoter sequence (pGL3/c-jun) or with a mutant form of c-jun promoter (pGL3/c-jun⫺). (Both are kindly provided by Drs. McCance and Baglia, University of Rochester, Rochester, NY.) A pCMV -galactosidase vector was used as internal control for transfection efficiency. Arsenite Treatment of the Cells—Arsenite (Sigma) was freshly prepared for each experiment in PBS and added to the cell culture at a final concentration of 5 or 10 M. Cells were treated for the durations indicated in appropriate figure legends. Thereafter, cells were harvested, washed with PBS twice, and then used for experiments. Cell Culture and Transfection of 293A Cells with hBVR and hHO-1 Expression Vectors—293A cell line (a human embryonic kidney cell line) was obtained from Invitrogen. Cells were grown in 10-cm plates with Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum and 1% penicillin-G/streptomycin for 24 h or until a 70% confluency was reached. Cells were subsequently transfected with 4 g of plasmid in each 10-cm dish using Transfectin Reagent (Bio-Rad), according to the manufacturer’s protocol. This protocol was found to be optimal for overexpression of hBVR and hHO-1, resulting in ⬃85% transfection efficiency as detected by fluorescent microscopy. 24 h after transfections, supernatant containing retrovirus expressing siRNA was added. This time point was designated as the “0” point. 4 and 16 h after the initial infection with retrovirus, additional virus was added at the same concentration. Samples were obtained at time points indicated in appropriate figure legends. 3 ⫻ 106 cells were used for RNA analysis and 1 ⫻ 106 for protein analysis. Isolation of Cytosolic Fraction—293 cells were washed with PBS and then homogenized in the extraction buffer (220 mM mannitol, 68 mM sucrose, 50 mM Pipes-KOH, 50 mM KCl, 5 mM EGTA, 2 mM MgCl2, and protease inhibitors). The homogenate was centrifuged at 400 ⫻ g at 4 °C for 10 min, and the supernatant fraction was further centrifuged at 10,000 ⫻ g for 10 min; the resultant supernatant was used as the cytosolic fraction for Western blot analysis of cytochrome c levels. The protein amount was determined with a Bradford protein assay. Western and Northern Blot Analyses—These analyses were carried out as before (33). Briefly, cells infected as above were collected, washed with PBS, resuspended in 1 ml of lysis buffer (100 mM Tris, 150 mM NaCl, 1 mM EDTA, 0.1% Nonidet P-40, 1 g/ml aprotinin, 1 g/ml pepstatin), and incubated at 4 °C for 1 h. Cell lysate was subjected to electrophoresis on 14% SDS-polyacrylamide gels and transferred to nitrocellulose membrane (Pall Corp., Ann Arbor, MI). hBVR and hHO-1 fusion proteins were detected using mouse anti-FLAG antibodies (1: 1000, v/v, Sigma) as the primary antibody and alkaline phosphataselabeled anti-mouse IgG (1:5000, v/v, Bio-Rad) as the secondary antibody. hBVR and hHO-1 proteins were visualized by nitro blue tetrazolium/5-bromo-4-chloro-3-indolyl phosphate staining (Calbiochem) or ECL system (Amersham Biosciences). Anti-mouse monoclonal anti-cytochrome c and rabbit polyclonal anti-TRAIL antibody were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-PARP antibody (C-2-10) was purchased from BioMol (Plymouth Meeting, PA). Total RNA was extracted with TRIzol Reagent (Invitrogen) from 293A cells. 10 g samples were subjected to denaturing formaldehyde gel electrophoresis and subsequently transferred to Hybond membrane. Membranes were probed with a full-length hBVR cDNA (48) or 0.8-kb fragment of hHO-1 cDNA (49). A 1.1-kb fragment of human -actin cDNA was used as internal control for loading. pCDNA3 containing DR-5 cDNA was cut by EcoRI/HindIII and used as probe for DR-5. Probes were labeled using [␣-32P]dCTP with the random primers labeling system (Invitrogen). Pre-hybridization, hybridization, and autoradiography were performed as described previously (50). ELISA—HO-1 protein was measured by using an ELISA kit developed by Stressgen Bioreagents (Victoria, British Columbia, Canada) according to the manufacturer’s instructions. Measurement of BVR Activity—Human 293A cells infected with pSuper-Retro-siBVR were lysed in buffer containing 50 mM Tris-HCl (pH 7.4), 75 mM NaCl, 20 mM MgCl2, 10 mM MnCl2, 1% Nonidet-P-40, 2 17086 siRNA-mediated Gene Silencing of Biliverdin Reductase mM EDTA, 2 mM EGTA, 10% glycerol, protease inhibitor mixture (1 g/ml aprotinin, 1 g/ml leupeptin, 1 g/ml pepstatin, and 0.1 mM phenylmethylsulfonyl fluoride), and phosphatase inhibitors (10 mM NaF, 1 mM NaVO4). Noninfected 293A cells were used as control. BVR activity was measured at pH 6.7 as described previously (51). The rate of conversion of biliverdin to bilirubin was determined as the increase in absorbance at 450 nm at 25 °C. Specific activity is expressed as nanomoles of bilirubin/min/mg of protein. Luciferase Assay—Transfection of 293A cells in 24-well plates was performed by using Lipofectamine (Invitrogen) with 0.4 g of plasmid containing PGL31 c-jun promoter fragment (⫺225 to ⫹150) (52), 0.4 g of plasmid containing c-jun promoter mutant construct (pGL3/c-Jun⫺) and containing mutated AP-1 site (53), 0.4 g of either empty pcDNA3, pCMV -galactosidase plasmid, or pGL3 vector (without the inserts). To test the role of BVR and HO-1 in arsenite-dependent c-jun induction, 293A cells were infected with retrovirus expression either siRNA for BVR or HO-1. After 24 h of infection, cells were treated with arsenite for an additional 8 h and then harvested, washed, and lysed. 5 l of lysate from each sample was used for luciferase assay using a Promega kit (Madison, WI). -Galactosidase activity was used to assess transfection efficiency, and luciferase activity was normalized against -galactosidase activity. In Vitro BVR Protein Translation, Nuclear Extraction, -Phosphatase Treatment, and Gel Mobility Shift Assay—293A cells were treated or untreated with arsenite. 107 cells were harvested at 8 or 24 h after treatment; cells were washed with cold PBS and then lysed with hypotonic buffer. Nuclei were collected by centrifugation and used for extract preparation. In vitro BVR protein translation was performed using a TNT Quick Coupled Translation System (Promega, Madison, WI). Briefly, full-length hBVR cDNA was cloned into a pcDNA3 expression vector downstream from the T7 RNA polymerase promoter. 2.0 g of recombinant plasmid DNA obtained was used for protein translation with TNT Quick Master Mix in a 50-l reaction volume for 90 min at 30 °C. To determine whether dephosphorylation of BVR impairs its binding to AP-1, the following experiment was carried out using -phosphatase (Sigma). 293A cells were transfected with pEGFP-FLAG-hBVR, using Lipofectamine Reagent (Bio-Rad). 24 h later, cells were treated with 5 M arsenite for 8 h. Nuclear extract was isolated and was subsequently treated with -phosphatase at concentrations of 20 – 80 units/20-l concentrations for 30 min at 37 °C. Thereafter, the samples were used for gel mobility shift assay for AP-1 binding. The in vitro translated protein and nuclear extract were analyzed in gel mobility shift assay for their DNA binding capability to an AP-1 consensus oligo (54). The sequences of oligonucleotides in double strands used in the present study were 5⬘-CGC TTG ATG AGT CAG CCG GAA CGC TTG ATG AGT CAG CCG GAA-3⬘. The oligos were labeled with [␥-32P]ATP by T4 kinase (Invitrogen). For DNA binding assay, 3 l of in vitro translated protein or nuclear extract was preincubated with 2 l of binding buffer (20% glycerol, 5 mM MgCl2, 2 mM EDTA, 2.5 mM dithiothreitol, 250 mM NaCl, and 50 mM Tris-HCl, 0.25 mg/ml poly(dI):poly(dC)) in an 8-l reaction volume for 10 min at room temperature. Then 2 l of labeled oligo was added, and samples were incubated for an additional 20 min at room temperature. The DNAprotein binding complexes were subjected to electrophoresis on a 6% nondenatured polyacrylamide gel and processed for autoradiography. In order to identify BVR in the DNA-protein complex, supershift experiments were performed using rabbit polyclonal anti-human BVR antibodies (55). Each experiment was repeated at least three times to ascertain the reproducibility of results. Detection of Apoptotic Cells—To detect apoptotic cells after arsenite treatment, 293A cells were treated with 10 M arsenite for 12 h and subsequently harvested. Cells were washed and fixed with 75% ethanol and stained with 20 M propidium iodide with 2 g/ml of RNase A. The cells were further analyzed by flow cytometry for apoptosis based on the propidium iodide staining. 103 cells were analyzed for each sample. RESULTS Arsenite Activates BVR Prior to Induction of HO-1 Expression—Previous studies (30) had shown that in the presence H2O2, BVR is activated and displays increased phosphorylation. To examine whether activation of BVR is relevant to HO-1 oxidative response to arsenite, 293A cells were treated with low arsenite concentrations (5 M), and in the following 8 h, BVR activity and HO-1 protein levels were monitored. ELISA was used for detection of HO-1 protein. As shown in Fig. 1, after the FIG. 1. Arsenite-mediated activation of BVR precedes induction of HO-1. 293A cells were treated with 5 M arsenite and harvested at the indicated time points for detection of BVR activity and HO-1 protein levels. BVR activity was analyzed at pH 6.7 with NADH as the cofactor. ELISA measured HO-1 protein levels. Data shown are mean ⫾ S.D. of three experiments. The experiments details are provided under “Materials and Methods.” treatment of the 293A cells with arsenite, a significant increase (4 –5-fold) of BVR activity was observed at 1 h after treatment. BVR activity gradually decreased thereafter and essentially returned to basal level 8 h later. There was no change in HO-1 protein levels in the 1st h after addition of arsenite. A significant increase in HO-1 level was observed 4 h after the treatment that further increased at the 8-h time point. Arsenite-mediated Increase in BVR Activity Is Not Accompanied by Induction in Its Expression but by Increase in Its Serine/ Threonine Phosphorylation—Fig. 2 shows that the increase in BVR activity was not essentially due to activation of gene expression, as indicated by the unchanged level of its 1.5-kb transcript detected by Northern blot analysis. A significant increase in HO-1 mRNA message, however, was observed 2 h after the addition of arsenite followed by robust increases at 4and 8-h time points. Arsenite is known to activate serine/threonine kinases (56). We have shown recently (22) that an increase in the phosphorylation of BVR triggers an increase in its reductase activity. To examine whether arsenite treatment also increases BVR phosphorylation, 293A cells were treated with 5 M arsenite for 30 or 60 min, lysed, and subjected to immunoprecipitation using anti-hBVR antibodies. The immunoprecipitations were used for Western blot analyses of phosphorylated BVR. For detection, a mixture of anti-serine and anti-threonine phosphoantibodies in a 1:1 ratio was used. In order to test whether an increase in BVR activity reflected an increase in BVR protein, cells were similarly treated with arsenite and harvested at 1, 2, 4, and 8 h after treatment, and cell lysate was used for detection of BVR protein levels by Western blotting. Findings of the study shown in Fig. 3 demonstrated that arsenite indeed induced a significant increase in BVR phosphorylation at 30 and 60 min after treatment (Fig. 3a), whereas total BVR protein levels were not affected by arsenite up to 8 h after treatment (Fig. 3b). Retroviral Vector Based siRNA System Effectively Knocks Down BVR mRNA in 293A Cells—The role of BVR in oxidative stress response of cells to arsenite was examined using a retroviral vector-based siRNA system to create a BVR knock-down model. 293A cells transfected with expression construct for EGFP-FLAG-BVR fusion proteins were infected with pSuperRetro-siBVR constructs and were harvested at the 8- and 24-h time points. Control cells were transfected with EGFP-FLAGBVR expression construct. The efficiency of the vector-based siRNA-mediated Gene Silencing of Biliverdin Reductase 17087 FIG. 2. Arsenite treatment increases HO-1 mRNA but not that of BVR. Total RNA was isolated from 293A cells treated with arsenite (5 M) for the indicated periods. Northern blotting analysis was performed as described in the text. Membrane was probed with an 800-bp hHO-1 cDNA fragment, together with the full-length hBVR cDNA. Each lane contained 10 g of RNA. The membrane was subsequently probed for -actin, used as a control for loading. FIG. 3. Serine/threonine phosphorylation of BVR is induced by arsenite treatment. 293A cells were transfected with pCDNA3FLAG-BVR expression vector. 24 h after transfection, cells were treated with 5 M arsenite for the indicated periods. Total cell lysate in RIPA buffer was subjected to immunoprecipitation with anti-FLAG antibodies. The precipitated proteins were separated on SDS-PAGE, transferred to polyvinylidene difluoride membrane, and probed with either anti-phosphoserine/-threonine antibodies (a) or with anti-hBVR antibodies (b). The ECL system was used for detection of signals. FIG. 4. Retroviral based siRNA system effectively knocked down BVR mRNA and protein levels in 293A cells. A pSuper-Retro viral based siRNA system was used to silence BVR mRNA in cells and to create an siRNA knock down 293A cell models. Experimental details are provided in the text. a, Northern blot analysis of BVR expression 24 h after infection with the inhibitory construct. The filter was reprobed for -actin that was used as control for RNA integrity and equal loading. b, Western blot analysis of BVR protein in 293A cells infected with BVR siRNA. The blot was first probed with anti-FLAG antibodies and re-probed with anti--actin antibodies, which served as control for loading. The bottom panel shows Coomassie staining of the same gel to evaluate the effect of siRNA on total cellular protein. c, fluorescence microscopy of the 293A cells transfected with EGFP-FLAG-hBVR or with the expression vector plus BVR siRNA. siRNA system to “knock down” BVR was assessed by Northern and Western blot analyses and by fluorescence microscopy. As shown in Fig. 4, BVR mRNA was nearly undetectable 24 h after infection (Fig. 4a). Western blot analysis indicated that BVR protein was also reduced to undetectable levels at this time point (Fig. 4b). Fig. 4c shows that 8 h after infection with pSuperRetro-siBVR, the EGFP-FLAG-BVR fluorescent fusion protein display was significantly attenuated when compared with control FIG. 5. BVR siRNA knocks down arsenite-dependent increases in BVR activity and HO-1 protein levels. Cells were seeded into 6-well plates and pretreated with retroviral construct containing BVR siRNA. 24 h later, cultures were treated with 5 M arsenite. Analysis of BVR activity and HO-1 protein levels were carried after 1 or 8 h. BVR activity is expressed as nanomoles of bilirubin/min/mg of total cell protein. ELISA was used to determine HO-1 protein levels. The results are expressed as average ⫾ S.D. of three experiments and normalized to total amount of proteins in each sample. *, p ⱕ 0.05 when compared with 0 time control. ⫹, p ⱕ when compared with appropriate arsenite-treated control. cells in the absence of infection with the inhibitory RNA. The experimental data indicated that the pSuper-Retro-siBVR system effectively decreased the target mRNA and thus could be used as an effective model for experiments described below to evaluate the function of BVR in cytoprotection and induction of HO-1 in 293A cells in response to arsenite treatment. BVR Influences Arsenite-dependent HO-1 Expression—BVR siRNA was used to test the role of BVR in HO-1 response to arsenite treatment. Cells were infected with retroviral vector expressing BVR siRNA for 24 h, followed by treatment with 5 M arsenite. BVR activity and HO-1 protein level were analyzed (Fig. 5a). As with the experimental results shown in Fig. 1, arsenite treatment significantly increased BVR at the 1-h time point, thereafter returning to the normal levels at 8 h. BVR siRNA treatment of the cells suppressed the arsenitemediated increase in BVR activity, as well as that of the control cells. The effect of siBVR treatment on HO-1 response to arsenite was examined. A robust and time-dependent increase in HO-1 protein was observed when measured at 8, 12, 16, and 24 h after arsenite treatment. This increase in HO-1 expression was suppressed by about 60% at 16 and 24 h when cells were pretreated with BVR siRNA. The finding that siBVR did not completely block the induction of HO-1 by arsenite is consistent with the reported involvement of a number of transcription factors in the induction of HO-1 that are activated by arsenite (44) and indicate that BVR is not the sole transcription factor for the HO-1 response to arsenite. Furthermore, the finding that BVR siRNA in the absence of arsenite exposure did not significantly affect basal levels of HO-1 suggests that BVR influences arsenite-driven HO-1 induction. Arsenite Treatment of 293A Cells Enhances Binding Ability of BVR Protein to AP-1 Regulatory Elements—To examine whether involvement of BVR in arsenite-mediated induction of 17088 siRNA-mediated Gene Silencing of Biliverdin Reductase FIG. 6. Arsenite treatment increases BVR-AP-1 binding that is phosphorylation-dependent. a, arsenite treatment increases BVRAP-1 binding. 293A cell were treated with 5 M arsenite for 8 or 24 h; nuclear extract was obtained and used for gel mobility shift assay for AP-1 binding in the presence and absence of anti-hBVR antibodies (supershift) (55). b, dephosphorylation of BVR by -phosphatase treatment abolishes BVR-AP-1 binding. Nuclear extract was isolated from the 293A cells transfected with pEGFP-BVR and treated with 5 M arsenite for 8 h. The extract was treated with -phosphatase for 30 min at 37 °C and subsequently used for gel mobility shift assay for AP-1 binding. Experimental procedures are described in detail in the text. FIG. 7. Biliverdin, but not bilirubin, impairs BVR-AP-1 complex formation. In vitro translated BVR was obtained using the TNT Quick Coupled Translation System (Promega, Madison, WI). The translated BVR protein was used for gel mobility shift assay. Left panel represents gel shift assay and shows that biliverdin inhibited BVR/AP-1 binding in vitro in a dose-dependent manner. Right panel represents one of the experiments to determine BVR/AP-1 binding in the presence of 60 M each of biliverdin and bilirubin. The DNA fragment (42 bp) was a synthesized oligo with two AP-1 sites (30). The experiment was repeated four times. t indicates in vitro translated BVR. HO-1 is accompanied by an increase in its AP-1 binding activity, gel mobility shift assay was performed at 8- and 24-h time points. Nuclear extract was prepared from 293A cells treated with 5 M arsenite and used in the assay for detection of protein binding to 32P-labeled AP-1 probe. Results are shown in Fig. 6. As shown in Fig. 6a, the nuclear extract obtained from the treated cells displayed an increased ability to form a protein-DNA complex. BVR was identified as a major component of the complex in the supershift assay using antibodies to hBVR. It is noteworthy that whereas activation of BVR, i.e. increased phosphorylation, is detected 1 h after arsenite treatment (Fig. 3), AP-1 binding remains in effect as late as 24 h. These observations are consistent with the likelihood of BVR functioning as a kinase at an early phase of cellular response to arsenite and as a transcription factor/DNA-binding protein at the later phase. To confirm further that increased BVR/AP-1 binding caused by arsenite treatment is related to phosphorylation of the protein, the effect of dephosphorylation of BVR on its DNA binding was examined. Previous studies had shown that treatment of hBVR with phosphatase effectively dephosphorylates the protein (22). Cells were transfected with pEGFP-FLAG-hBVR expression construct, followed by treatment with 5 M arsenite. 8 h later nuclear extract was obtained and treated with -phosphatase at 20 – 80 units/20 l of concentration for 30 min at 37 °C. Thereafter, samples were analyzed for AP-1 binding. As shown in Fig. 6b, dephosphorylation of BVR significantly reduced its AP-1 binding capability, and the decrease in the formation of BVR-AP-1binding complex was concentration-dependent. Biliverdin, but Not Bilirubin, Is an Effective Inhibitor of BVRAP-1 Binding in Vitro—Previous studies had shown that BVR is inhibited by bilirubin, whereas biliverdin is an effective inhibitor of HO-1 activity (1, 18). To examine whether this inhibition involves interference of BVR substrate with its DNA binding, TNT in vitro protein translation system was used to test BVRAP-1 complex formation in the presence or absence of biliverdin. Bilirubin was used as the control. The newly translated BVR protein was incubated with the 32P-labeled DNA fragment, containing 2 AP-1 sites, in the presence of either biliverdin or bilirubin. The results of gel mobility shift assay are shown in Fig. 7. In the presence of biliverdin, there was a notable decrease in signal for BVR-DNA complex formation. Bilirubin, however, did not affect formation of the complex. The result defines biliverdin as an effective inhibitor of BVR-AP-1 complex formation, an effect that was not shown by its reduced form. BVR Regulates Arsenite-mediated c-jun Promoter Activation—BVR-AP-1 complex formation using an adenovirus construct of hBVR to overexpress the protein was shown previously (33). BVR in a homodimeric form binds to DNA containing two AP-1 sites but not to a fragment containing a single AP-1 site (30). In order to establish involvement of BVR in AP-1 activation by arsenite, a pGL3 vector containing the c-jun promoter was used as a luciferase reporter system, where the c-jun promoter with two AP-1 sites was placed downstream from the luciferase reporter. Cells were co-transfected with siBVR, whereas siHO-1-infected cells were used as a control. A second control was cell-transfected with TRRE site-mutated c-jun promoter to address the specificity of the c-jun promoter. Data are shown in Fig. 8a. 293A cells transfected with the pGL3 vector containing the c-jun promoter showed a base-line luciferase activity. After treatment of the cells with 5 M arsenite, the luciferase activity of the c-jun promoter was increased approximately 3-fold. The same treatment, however, did not increase TRRE site-mutated c-jun promoter. When cells transfected with pGL3/c-jun promoter were pretreated with BVR siRNA and followed by arsenite treatment, luciferase activity was essentially similar to that of the untreated controls (PGL3/c-jun). Pretreatment of the cells with HO-1 siRNA followed by arsenite treatment did not significantly affect lucif- siRNA-mediated Gene Silencing of Biliverdin Reductase FIG. 8. Knock down of BVR but not that of HO-1 prevents activation of c-jun promoter by arsenite. a, c-jun promoter reporter assay. 293A cells seeded into 24-well plates were transfected with either c-jun luciferase promoter reporter construct (pGL3/c-jun, 0.4 g) or with c-jun luciferase promoter reporter mutant construct (pGL3mt/ c-jun, 0.4 g) cells were cotransfected with pCMV -galactosidase (0.3 g) vector used as a control for normalization of transfection. 24 h after transfection cells were synchronized overnight by replacing growth medium with one containing 0.1% fetal bovine serum. Cells were treated with either siRNA for BVR or siRNA for HO-1 as described under “Materials and Methods.” After 24 h, cells were treated with 5 M arsenite. 8 h later, they were harvested, washed with PBS, lysed in reporter lysis buffer, and assayed for luciferase and -galactosidase activities. Luciferase activity normalized for transfection efficiency against -galactosidase activity is presented as mean ⫾ S.D. for three experiments with quadruplicate samples. b, Western blot analysis of HO-1 protein in 293A cells transfected with EGFP-FLAG-hHO-1 expression construct and infected with HO-1 siRNA. Total protein stained by Coomassie Blue was used as control for loading. c, 293A cells transfected with expression vector for EGFP-FLAG-HO-1 plus or minus HO-1 siRNA. EGFP fluorescence was detected by a microscope. erase activity. These observations were supportive of BVR involvement of HO-1 response to arsenite through its AP-1 binding capability. To confirm that HO-1 siRNA indeed had effectively knocked down HO-1 protein, cells were infected with the EGFP-HO-1 expression construct, and 24 h later they were infected with siHO-1 for an additional 24 h. Cell lysate was prepared and used for the detection of protein levels by Western blotting (Fig. 8b). Fluorescent microscopy was used to visualize HO-1 protein in intact cells (Fig. 8c). As noted, the HO-1 siRNA infection was highly effective in attenuating HO-1 levels, thus generating a useful model for further experiments described below. siRNA-mediated Knock Down of BVR Increases Sensitivity of 293A Cells to Arsenite-mediated Apoptotic Cell Death—To evaluate whether BVR itself, independent of HO-1, is an effective cytoprotector, arsenite-mediated apoptosis by a high concentration of the compound was examined. 293A cells were infected with BVR siRNA or HO-1 siRNA retrovirus for 24 h, followed by treatment with 10 M arsenite. 12 h later, cells were harvested, washed, fixed with 75% ethanol, and stained with 20 M propidium iodide. The cells were analyzed by flow cytometry for apoptosis based on propidium iodide staining. As shown in Fig. 9, BVR siRNA-mediated knock down in 293A cells resulted in a 4-fold increase in the number of apoptotic cells when compared with control. Most surprisingly, in the 293A cells infected with HO-1 siRNA retrovirus, a significant effect was not observed after arsenite treatment when compared with control cells. The data reveal a previously unknown function of BVR as an effec- 17089 tive protector of cells against oxidative stress and suggest independence of this activity from that of reduction of biliverdin and production of bilirubin. To investigate the mechanism by which siBVR potentiates arsenite-mediated apoptosis, 293 cells were depleted of hBVR using siBVR prior to treatment, and the effects of modulation of BVR on key factors involved in apoptosis were examined (Fig 10). An early event in apoptotic signaling cascade is the release of cytochrome c from mitochondria, which precedes activation of caspases, FasL-induced cleavage of poly(ADP-ribose) polymerase (PARP), followed by DNA fragmentation. Activation of caspase-3 by TRAIL/Apo-2L, a member of tumor necrosis factor superfamily, is triggered through its binding to death receptors DR4 and DR5. To examine effect of BVR knock down on arsenite-mediated cytochrome c release, cells infected with siBVR were treated with 10 M arsenite, lysed 24 h after the treatment, and levels of cytochrome c in the cytosol were examined by immunoblotting. Control cells were not infected with the interfering RNA but received the same regimen of arsenite treatment (Fig. 10a). We observed an increase in cytochrome c levels in the cytosolic fraction of arsenite-treated cells when compared with control cells. This increase was augmented when cells were pre-exposed to BVR siRNA. When the total cell lysate post-arsenite treatment was examined for the appearance of the 85-kDa cleavage product of a 116-kDa PARP was detected by Western blot analysis in both control and siRNAtreated cells (Fig. 10a). The appearance of the cleaved product was more pronounced in siRNA-treated cells. In cells transfected with wtBVR, the 85-kDa cleavage product was essentially undetectable. To investigate further the contribution of BVR to anti-apoptotic events, the effect of transfection of cells with wtBVR or siBVR on another pro-apoptotic factor TRAIL was assessed. As noted in Fig. 10a, similar to PARP, the marked induction of TRAIL by arsenite treatment was attenuated by overexpression of wtBVR and conversely increased by depletion of endogenous BVR (Fig. 10a). Binding of TRAIL to death receptor DR5 is essential for the induction of TRAIL signaling of apoptotic cell death. TRAIL triggers apoptosis through death receptors DR4 and DR5; DR5 is a greater contributor to apoptotic signaling. Therefore, whether modulation of cellular levels of BVR would affect arsenite dependent DR-5 induction was examined. As shown in Fig. 10b, a modest induction of DR-5 mRNA was observed at 12, 16, and 24 h after treatment of control cells with arsenite. However, when cells were pre-treated with BVR siRNA, a robust induction of DR-5 mRNA upon treatment with arsenite was observed. The arsenite-dependent DR-5 induction was again attenuated when cells were transfected with wtBVR. Treatment of cells with empty virus did not have a significant effect on DR-5 mRNA levels. The results indicate that the presence of BVR is important in preventing induction of DR-5 mRNA in events that culminate in apoptosis. Together, the findings of this series of experiments suggest that BVR prevents cells from proceeding to apoptosis by interfering at several points along the cell death pathway. DISCUSSION The findings of this study suggest that BVR has a prominent and multidimensional role in the regulation of HO-1 stress response. Furthermore, it identifies the reductase, independent of bile pigments, as an essential component of cytoprotection against oxidative challenge. In turn, these findings reflect the collective functions of BVR as a reductase, with the ability to produce bilirubin and inactivate biliverdin, and as a kinase and component of cell signaling pathways, with the ability to influence gene expression. 17090 siRNA-mediated Gene Silencing of Biliverdin Reductase FIG. 9. BVR knock down, but not that of HO-1, decreases viability of 293A cells treated by arsenite. 293A cells were infected with BVR siRNA or HO-1 siRNA vectors. 24 h later, cells were exposed to 10 M arsenite, and after 12 h, cells were analyzed for apoptosis by flow cytometry based on propidium iodide staining of DNA content. Upper panel, the set of histograms shows the effect of siBVR and siHO-1 on the number of apoptotic cells. Lower panel, bar graph represents statistical analysis of apoptosis assay data of cells treated with arsenite. Data shown are mean ⫾ S.D. of five experiments. Although the heme molecule can be degraded to pyrrolic complexes in vivo and by chemical reactions in vitro, the HO/ BVR pathway is the only effective mechanism for production of CO, biliverdin IX␣, and bilirubin IX␣ (11). Inference to the role of the HO/BVR system in cell signaling and in cytoprotection is based on the current understanding of biological activities of the heme degradation products by this combination of enzymes. The reduction product of BVR activity is an effective intracellular antioxidant (57) and together with CO modulates immune effector functions (12–15, 19). The substrate for BVR, biliverdin IX␣, is an effective modulator of signal transduction pathways and inhibits protein phosphorylation and kinase activity (17, 58 – 60). Direct evidence for the role of biliverdin in cell signaling pathways was offered by the identification of biliverdin IX␣ as the determinant factor in the dorsal axis development in Xenopus laevis embryos (61). The observation that biliverdin attenuates BVR-DNA complex formation (Fig. 7) is a novel finding and further supports a role for the tetrapyrrole in cell signaling and identifies biliverdin as one component of the multidimensional mechanisms by which BVR regulates HO-1 expression. In this context, the reductase activity of BVR would serve as the means to inactivate the inhibitor of its binding to AP-1 sites in the HO-1 promoter. Biliverdin-mediated impairment of AP-1-BVR complex formation is consistent with the earlier work that had shown inhibition of HO activity in vivo by the tetrapyrrole (18). In fact, with the exception of biliverdin, to date no other heme pathway products or normal cell constituents have been found to inhibit HO activity in vivo or in vitro. The occurrence of a regulatory loop in the cell to control the heme degradation process, which would involve a combination of inhibitory effects of biliverdin on HO-1 expression and its catalytic activity, and activation of BVR by oxidant inducers of HO-1 can be considered. In this scenario, activation of BVR and rapid conversion of biliverdin to bilirubin would allow for derepression of HO-1 expression and increased activity; product (bilirubin) inhibition of BVR activity then would allow for buildup of biliverdin levels; this in turn, would cause product (biliverdin) inhibition of the oxygenase, hence permitting return to normal of the heme degradation process. The various components of this potential loop have been demonstrated here as well as in previous reports. For instance, inhibition of BVR activity by bilirubin and the initial suppression of HO activity in vivo by biliverdin followed by a rebound increase were reported long ago (1, 18). The inhibitory effect of biliverdin on BVR/AP-1 was documented in the present study. The concentration of biliverdin used for in vivo experiments (18) was comparable with that employed in the gel mobility shift assay (Fig. 7). It is relevant to note that exceedingly high concentrations of biliverdin can accumulate in the cell as the consequence of exposure to compounds that disrupt cell signaling pathways (58). A wide range of functions in the cell are subject to a change in activity of HO-1; this includes those that are controlled by NO radicals (16, 19, 20, 62– 65). Therefore, interplay between BVR, its substrate, and its product in the regulation of the stress response of HO-1 would have a major impact on the cell. However, the mechanism for cytoprotection offered by BVR as indicated by the striking increase in the number of apoptotic cells when cells infected with siBVR are exposed to arsenite (Fig. 9) is likely not to involve inhibition of bilirubin formation and exacerbation of damaging effects of oxygen free radicals siRNA-mediated Gene Silencing of Biliverdin Reductase FIG. 10. BVR attenuates arsenite-mediated apoptosis by interfering with cytochrome c and TRAIL-dependent apoptotic pathway. a, Western blot analysis effect of BVR knock down on cytochrome c, TRAIL, and PARP. Cells were either transfected with wild type hBVR expression construct (pcDNA3-BVR) or infected with construct containing siRNA for BVR. Transfected cells treated with 10 M As for 24 h were used for preparation of total cell lysate and the cytosolic fractionation. The cytosolic fraction was subjected to 15% SDS-PAGE, and the blotted proteins were used for detection of cytochrome c using monoclonal antibodies to the protein followed by the ECL detection system. Total cell lysate was used for immunoblot analysis of PARP and TRAIL using appropriate antibodies and the ECL detection system. Intact and cleaved PARP proteins are indicated as 116- and 85-kDa bands, respectively. Same blot was stripped and probed with anti-actin antibodies used as a control for loading. b, Northern blot analysis of DR5 levels in cells treated with arsenite in the presence of increased or knocked down levels of BVR expression. Cells transfected with wtBVR or infected with BVR siRNA were treated for the indicated periods with 10 M arsenite. Total RNA was isolated and used for Northern blot analysis. The membrane was probed with cDNA for DR5. Equal loading is indicated in the lower panel showing 28 S and 18 S RNA. The above series of experiments were performed twice. Experimental details are provided under “Materials and Methods.” (66). Rather, this element of the BVR role in cytoprotection is clearly independent of HO-1 expression and must involve the kinase activity of BVR and its input into the signal transduction pathways. Kinases control a wide variety of cellular functions, including apoptosis. Previous work with gene array analysis had identified several members of the signal transduction pathways as potential targets for regulation by BVR. This includes HSF1 (heat shock transcription factor1), heat shock proteins 90 and 27, Bcl-2, Cox-2, protein kinase C␣, and WISP3 (inducible signaling pathway protein 3). Among the list were ATF-2 (CREB-2) and c-jun (33). The finding that the c-jun promoter activation by arsenite can be blocked when BVR expression is knocked down (Fig. 8) places the reductase in the position of having a direct role in influencing not only HO-1 gene expression but rather influencing that of an extended list of stressresponsive genes. In the case of c-Jun, because it can form a DNA complex as a homodimer or a heterodimer not only with c-Fos but also with ATF-2/CREB (54, 67, 68), a change in the expression and activation state of c-Jun predictably would alter the profile of the cell signaling pathways. The fact that the knock down of BVR augmented cell killing activity of arsenite 17091 is clearly consistent with the role of the reductase in the expression of genes that affect cell growth and proliferation. Among those, the ATF-2/CREB and mitogen-activated protein kinase pathways are the most notable because of their input in the response of the cell to growth factors, mitogens and cytokines. The unexpected finding that attenuation of BVR expression markedly increased the proportion of apoptotic cells in arsenite-treated preparations and increased the levels of factors associated with the apoptotic mode of cell death, including cytochrome c, TRAIL, death receptor-5 (DR-5) mRNA, and PARP, further supports the suggestion that the role of BVR in cytoprotection against oxidants extends beyond regulating bilirubin and CO production by HO-1. We were indeed surprised by the observation that siHO-1 did not influence arseniteinduced apoptosis in 293A cells. This was particularly unexpected considering a recent report on the enhancement of ischemia/reperfusion-induced lung injury caused by introduction of the lung-specific siHO-1 (69). The divergence of two sets of findings is perplexing because in the present study we carefully established that in fact the HO-1 siRNA construct was effective at suppressing both the HO-1 protein and its RNA. Accordingly, the only explanation that can be offered at this time is activation of different pathways of signaling by arsenite and ischemia/reperfusion. Arsenite induces production of oxygen radicals, whereas ischemia/reperfusion injury has the initial hypoxic component. Thus, the profile of genes activated by arsenite and ischemia/reperfusion could differentially affect pathways of cell death and survival. The differences in the model system and constructs are also among the possible explanations. Based on the data and reasoning offered in this report, it is not unreasonable to suggest that experimental modulation of BVR in the cell could be used to enhance or attenuate the response of the cell to external stimuli. The observation that by silencing BVR expression the cell killing activity of arsenite is enhanced is a strong argument in support of this suggestion. Arsenite is an established human carcinogen that causes chromosomal aberrations, sister chromatid exchange, and micronuclei formation (47). Although the mechanism of its carcinogenicity is poorly understood, a substantial amount of evidence points to the role of hydroxy radicals, the activator of BVR (22), in the genotoxicity (70, 71). Because oxygen radicals are suspected to underlie the etiology of a long list of pathophysiological conditions, it would be reasonable to suggest that BVR with its multidimensional input into the regulation of oxidative stress-responsive genes may find a useful place in therapeutic settings. Acknowledgments—We are grateful to W. El-Deiry (University of Pennsylvania, Philadelphia) for the gift of DR-5 cDNA; J. Boyce for the preparation of the manuscript, and J. Smith for illustrations. REFERENCES 1. Kutty, R. K., and Maines, M. D. (1981) J. Biol. Chem. 256, 3956 –3962 2. Singleton, J. W., and Laster, L. (1965) J. Biol. Chem. 240, 4780 – 4789 3. Chikuba, K., Yubisui, T., Shirabe, K., and Takeshita, M. (1994) Biochem. Biophys. Res. Commun. 198, 1170 –1176 4. Komuro, A., Tobe, T., Hashimoto, K., Nakano, Y., Yamaguchi, T., Nakajima, H., and Tomita, M. (1996) Biol. & Pharm. Bull. 19, 796 – 804 5. Shalloe, F., Elliott, G., Ennis, O., and Mantle, T. J. (1996) Biochem. J. 316, 385–387 6. Yamaguchi, T., Komoda, Y., and Nakajima, H. (1994) J. Biol. Chem. 269, 24343–24348 7. Maines, M. D. (1988) FASEB J. 2, 2557–2568 8. Shibahara, S., Muller, R. M., and Taguchi, H. (1987) J. Biol. Chem. 262, 12889 –12892 9. Keyse, S. M., and Tyrrell, R. M. (1989) Proc. Natl. Acad. Sci. U. S. A. 86, 99 –103 10. Ewing, J. F., and Maines, M. D. (1991) Proc. Natl. Acad. Sci. U. S. A. 88, 5364 –5368 11. Maines, M. D. (1992) Heme Oxygenase: Clinical Applications and Functions, pp. 1–276, CRC Press, Inc., Boca Raton, FL 12. Haga, Y., Tempero, M. A., and Zetterman, R. K. (1996) Biochim. Biophys. Acta 1316, 29 –34 17092 siRNA-mediated Gene Silencing of Biliverdin Reductase 13. Willis, D., Moore, A. R., Frederick, R., and Willoughby, D. A. (1996) Nat. Med. 2, 87–90 14. Nakagami, T., Toyomura, K., Kinoshita, T., and Morisawa, S. (1993) Biochim. Biophys. Acta 1158, 189 –193 15. Parry, N., Buelow, R., Jiang, J., Garcia, B., and Zhong, R. (1999) Transplantation 67, 5252–5258 16. Otterbein, L. E., Soares, M. P., Yamashita, K., and Bach, F. H. (2003) Trends Immunol. 24, 449 – 455 17. Hansen, T. W., Mathiesen, S. B., and Walaas, S. I. (1996) Pediatr. Res. 39, 1072–1077 18. Kutty, R. K., and Maines, M. D. (1984) Biochem. Biophys. Res. Commun. 122, 40 – 46 19. Otterbein, L. E., Bach, F. H., Alam, J., Soares, M., Tao Lu, H., Wysk, M., Davis, R. J., Flavell, R. A., and Choi, A. M. (2000) Nat. Med. 6, 422– 428 20. Morse, D., Sethi, J., and Choi, A. M. (2002) Crit. Care Med. 30, S12–S17 21. Song, R., Mahidhara, R. S., Zhou, Z., Hoffman, R. A., Seol, D. W., Flavell, R. A., Billiar, T. R., Otterbein, L. E., and Choi, A. M. (2004) J. Immunol. 172, 1220 –1226 22. Salim, M., Brown-Kipphut, B. A., and Maines, M. D. (2001) J. Biol. Chem. 276, 10929 –10934 23. van Straaten, F., Muller, R., Curran, T., Van Beveren, C., and Verma, I. M. (1983) Proc. Natl. Acad. Sci. U. S. A. 80, 3183–3187 24. Gazin, C., Dupont de Dinechin, S., Hampe, A., Masson, J. M., Martin, P., Stehelin, D., and Galibert, F. (1984) EMBO J. 3, 383–387 25. Hinnebusch, A. G. (1984) Proc. Natl. Acad. Sci. U. S. A. 81, 6442– 6446 26. Bohmann, D., Bos, T. J., Admon, A., Nishimura, T., Vogt, P. K., and Tjian, R. (1987) Science 238, 1386 –1392 27. Hoeffler, J. P., Meyer, T. E., Yun, Y., Jameson, J. L., and Habener, J. F. (1988) Science 242, 1430 –1433 28. Moye-Rowley, W. S., Harshman, K. D., and Parker, C. S. (1989) Genes Dev. 3, 283–292 29. Ishii, T., Itoh, K., Takahashi, S., Sato, H., Yanagawa, T., Katoh, Y., Bannai, S., and Yamamoto, M. (2000) J. Biol. Chem. 275, 16023–16029 30. Ahmad, Z., Salim, M., and Maines, M. D. (2002) J. Biol. Chem. 277, 9226 –9232 31. Kitamuro, T., Takahashi, K., Ogawa, K., Udono-Fujimori, R., Takeda, K., Furuyama, K., Nakayama, M., Sun, J., Fujita, H., Hida, W., Hattori, T., Shirato, K., Igarashi, K., and Shibahara, S. (2003) J. Biol. Chem. 278, 9125–9133 32. Li, N., Alam, J., Venkatesan, M. I., Eiguren-Fernandez, A., Schmitz, D., Di Stefano, E., Slaughter, N., Killeen, E., Wang, X., Huang, A., Wang, M., Miguel, A. H., Cho, A., Sioutas, C., and Nel, A. E. (2004) J. Immunol. 173, 3467–3481 33. Kravets, A., Hu, Z., Miralem, T., Torno, M. D., and Maines, M. D. (2004) J. Biol. Chem. 279, 19916 –19923 34. Maines, M. D., Ewing, J. F., Huang, T. J., and Panahian, N. (2001) J. Pharmacol. Exp. Ther. 296, 1091–1097 35. Barchowsky, A., Dudek, E. J., Treadwell, M. D., and Wetterhahn, K. E. (1996) Free Radic. Biol. Med. 21, 783–790 36. Chen, N. Y., Ma, W. Y., Huang, C., Ding, M., and Dong, Z. (2000) J. Environ. Pathol. Toxicol. Oncol. 19, 297–305 37. Shen, Z. Y., Shen, W. Y., Chen, M. H., Shen, J., Cai, W. J., and Yi, Z. (2002) World J. Gastroenterol. 8, 40 – 43 38. Jung, D. K., Bae, G. U., Kim, Y. K., Han, S. H., Choi, W. S., Kang, H., Seo, D. W., Lee, H. Y., Cho, E. J., Lee, H. W., and Han, J. W. (2003) Exp. Cell Res. 290, 144 –154 39. Liao, W. T., Chang, K. L., Yu, C. L., Chen, G. S., Chang, L. W., and Yu, H. S. (2004) J. Investig. Dermatol. 122, 125–129 40. Wijayanti, N., Huber, S., Samoylenko, A., Kietzmann, T., and Immenschuh, S. (2004) Antioxid. Redox Signal 6, 802– 810 41. Alam, J. (1994) J. Biol. Chem. 269, 25049 –25056 42. Choi, A. M., and Alam, J. (1996) Am. J. Respir. Cell Mol. Biol. 15, 9 –19 43. Elbirt, K. K., Whitmarsh, A. J., Davis, R. J., and Bonkovsky, H. L. (1998) J. Biol. Chem. 273, 8922– 8931 44. Gong, P., Stewart, D., Hu, B., Vinson, C., and Alam, J. (2002) Arch. Biochem. Biophys. 405, 265–274 45. Kietzmann, T., Samoylenko, A., and Immenschuh, S. (2003) J. Biol. Chem. 278, 17927–17936 46. Cavigelli, M., Li, W. W., Lin, A., Su, B., Yoshioka, K., and Karin, M. (1996) EMBO J. 15, 6269 – 6279 47. Bode, A. M., and Dong, Z. (2002) Crit. Rev. Oncol. Hematol. 42, 5–24 48. Maines, M. D., Polevoda, B. V., Huang, T. J., and McCoubrey, W. K., Jr. (1996) Eur. J. Biochem. 235, 372–381 49. Yoshida, T., Biro, P., Cohen, T., Muller, R. M., and Shibahara, S. (1988) Eur. J. Biochem. 171, 457– 461 50. Miralem, T., and Avraham, H. K. (2003) Mol. Cell. Biol. 23, 579 –593 51. Huang, T. J., Trakshel, G. M., and Maines, M. D. (1989) J. Biol. Chem. 264, 7844 –7849 52. Clarke, N., Arenzana, N., Hai, T., Minden, A., and Prywes, R. (1998) Mol. Cell. Biol. 18, 1065–1073 53. Han, T. H., Lamph, W. W., and Prywes, R. (1992) Mol. Cell. Biol. 12, 4472– 4477 54. Angel, P., and Karin, M. (1991) Biochim. Biophys. Acta 1072, 129 –157 55. Maines, M. D., and Trakshel, G. M. (1993) Arch. Biochem. Biophys. 300, 320 –326 56. Chen, W., Martindale, J. L., Holbrook, N. J., and Liu, Y. (1998) Mol. Cell. Biol. 18, 5178 –5188 57. Stocker, R. (2004) Antioxid. Redox Signal 6, 841– 849 58. Maines, M. D. (2003) Toxicol. Sci. 71, 9 –10 59. Phelan, D., Winter, G. M., Rogers, W. J., Lam, J. C., and Denison, M. S. (1998) Arch. Biochem. Biophys. 357, 155–163 60. Sinal, C. J., and Bend, J. R. (1997) Mol. Pharmacol. 52, 590 –599 61. Falchuk, K. H., Contin, J. M., Dziedzic, T. S., Feng, Z., French, T. C., Heffron, G. J., and Montorzi, M. (2002) Proc. Natl. Acad. Sci. U. S. A. 99, 251–256 62. Maines, M. D. (1997) Annu. Rev. Pharmacol. Toxicol. 37, 517–554 63. Motterlini, R., Foresti, R., Bassi, R., and Green, C. J. (2000) Free Radic. Biol. Med. 28, 1303–1312 64. Durante, W., Christodoulides, N., Cheng, K., Peyton, K. J., Sunahara, R. K., and Schafer, A. I. (1997) Am. J. Physiol. 273, H317–H323 65. Chen, Y. H., Yet, S. F., and Perrella, M. A. (2003) Exp. Biol. Med. 228, 447– 453 66. Baranano, D. E., Rao, M., Ferris, C. D., and Snyder, S. H. (2002) Proc. Natl. Acad. Sci. U. S. A. 99, 16093–16098 67. Ransone, L. J., Visvader, J., Sassone-Corsi, P., and Verma, I. M. (1989) Genes Dev. 3, 770 –781 68. Herdegen, T., and Leah, J. D. (1998) Brain Res. Brain Res. Rev. 28, 370 – 490 69. Zhang, X., Shan, P., Jiang, D., Noble, P. W., Abraham, N. G., Kappas, A., and Lee, P. J. (2004) J. Biol. Chem. 279, 10677–10684 70. Liu, S. X., Athar, M., Lippai, I., Waldren, C., and Hei, T. K. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 1643–1648 71. Qian, Y., Castranova, V., and Shi, X. (2003) J. Inorg. Biochem. 96, 271–278
© Copyright 2025 Paperzz