101
|
Wellmann S, Bührer C, Moderegger E, Zelmer A, Kirschner R, Koehne P, Fujita J, Seeger K. Oxygen-regulated expression of the RNA-binding proteins RBM3 and CIRP by a HIF-1-independent mechanism. J Cell Sci 2004; 117:1785-94. [PMID: 15075239 DOI: 10.1242/jcs.01026] [Citation(s) in RCA: 201] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The transcriptional regulation of several dozen genes in response to low oxygen tension is mediated by hypoxia-inducible factor 1 (HIF-1), a heterodimeric protein composed of two subunits, HIF-1alpha and HIF-1beta. In the HIF-1alpha-deficient human leukemic cell line, Z-33, exposed to mild (8% O(2)) or severe (1% O(2)) hypoxia, we found significant upregulation of two related heterogenous nuclear ribonucleoproteins, RNA-binding motif protein 3 (RBM3) and cold inducible RNA-binding protein (CIRP), which are highly conserved cold stress proteins with RNA-binding properties. Hypoxia also induced upregulation of RBM3 and CIRP in the murine HIF-1beta-deficient cell line, Hepa-1 c4. In various HIF-1 competent cells, RBM3 and CIRP were induced by moderate hypothermia (32 degrees C) but hypothermia was ineffective in increasing HIF-1alpha or vascular endothelial growth factor (VEGF), a known HIF-1 target. In contrast, iron chelators induced VEGF but not RBM3 or CIRP. The RBM3 and CIRP mRNA increase after hypoxia was inhibited by actinomycin-D, and in vitro nuclear run-on assays demonstrated specific increases in RBM3 and CIRP mRNA after hypoxia, which suggests that regulation takes place at the level of gene transcription. Hypoxia-induced RBM3 or CIRP transcription was inhibited by the respiratory chain inhibitors NaN(3) and cyanide in a dose-dependent fashion. However, cells depleted of mitochondria were still able to upregulate RBM3 and CIRP in response to hypoxia. Thus, RBM3 and CIRP are adaptatively expressed in response to hypoxia by a mechanism that involves neither HIF-1 nor mitochondria.
Collapse
Affiliation(s)
- Sven Wellmann
- Department of Pediatric Oncology/Hematology, Charité Campus Virchow-Klinikum, Medical University of Berlin, 13353 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Wang J, Wei Q, Wang CY, Hill WD, Hess DC, Dong Z. Minocycline up-regulates Bcl-2 and protects against cell death in mitochondria. J Biol Chem 2004; 279:19948-54. [PMID: 15004018 DOI: 10.1074/jbc.m313629200] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Robust neuroprotective effects have been shown for minocycline. Whether it also protects nonneuronal cells or tissues is unknown. More importantly, the mechanisms of minocycline protection appear multifaceted and remain to be clarified. Here we show that minocycline can protect kidney epithelial cells in vitro and protect the kidneys from ischemic injury in vivo. We further show that Bcl-2 is a key molecular determinant of minocycline protection. Minocycline protected kidney epithelial cells against apoptosis induced by hypoxia, azide, cisplatin, and staurosporine. The protection occurred at mitochondria, involving the suppression of Bax accumulation, outer membrane damage, and cytochrome c release. Minocycline induced Bcl-2, which accumulated in mitochondria and interacted with death-promoting molecules including Bax, Bak, and Bid. Down-regulation of Bcl-2 by specific antisense oligonucleotides abolished the cytoprotective effects of minocycline. Thus, minocycline can protect neuronal as well as nonneuronal cells and tissues. One mechanism for minocycline protection involves the induction of Bcl-2, an antiapoptotic protein.
Collapse
Affiliation(s)
- Jinzhao Wang
- Department of Cell Biology and Anatomy, Medical College of Georgia, 1459 Laney Walker Boulevard, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
103
|
Pichiule P, Chavez JC, LaManna JC. Hypoxic Regulation of Angiopoietin-2 Expression in Endothelial Cells. J Biol Chem 2004; 279:12171-80. [PMID: 14702352 DOI: 10.1074/jbc.m305146200] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure of endothelial cells to hypoxia-induced angiopoietin-2 (Ang2) expression. The increase in Ang2 mRNA levels occurred by transcriptional regulation and by post-transcriptional increase in mRNA stability. Induction of Ang2 mRNA resulted in an increase of intracellular and secreted Ang2 protein levels. Since the transcriptional regulation of several genes involved in angiogenesis during hypoxia is mediated by hypoxia-inducible factor-1 (HIF-1), it was conceivable that Ang2 expression might be regulated by the same oxygen-dependent mechanism. However, our data showed that pharmacological HIF inducers, CoCl(2) and DFO, did not affect Ang2 expression. Moreover, HIF-1-deficient hepatoma cell (Hepa1 c4) and its wild-type counterpart (Hepa1 c1c4) up-regulates Ang2 during hypoxia. These results indicated that hypoxia-driven Ang2 expression may be independent of the HIF pathway. Using neutralizing VEGF antibody or pharmacological inhibitors of VEGF receptors, we showed that hypoxia-induced VEGF participates but could not account completely for Ang2 expression during hypoxia. In addition, hypoxia elicited an increase of cyclooxygenase-2 (COX-2) expression and a parallel increase in prostanglandin E(2) (PGE(2)) and prostacyclin (PGI(2)) production. COX-2 inhibitors decreased the hypoxic induction of Ang2 and the hypoxic induction of PGE(2) and PGI(2) in a dose-dependent manner. Similarly, COX-2 but not COX-1 antisense treatment decreased hypoxic induction of Ang2 expression, and this effect was reversed by exogenous PGE(2). Finally, exogenous PGE(2) and PGI(2) were able to stimulate Ang2 under normoxic conditions. These findings suggest that COX-2-dependent prostanoids may play an important role in the regulation of hypoxia-induced Ang2 expression.
Collapse
MESH Headings
- Angiopoietin-2/biosynthesis
- Blotting, Western
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Cell Survival
- Cells, Cultured
- Cobalt/pharmacology
- Cyclooxygenase 1
- Cyclooxygenase 2
- Cyclooxygenase 2 Inhibitors
- Cyclooxygenase Inhibitors/pharmacology
- DNA, Complementary/metabolism
- DNA-Binding Proteins/metabolism
- Deferoxamine/pharmacology
- Dinoprostone/metabolism
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/metabolism
- Enzyme-Linked Immunosorbent Assay
- Epoprostenol/metabolism
- Humans
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1
- Hypoxia-Inducible Factor 1, alpha Subunit
- Inhibitory Concentration 50
- Iron Chelating Agents/pharmacology
- Isoenzymes/biosynthesis
- Lactones/pharmacology
- Membrane Proteins
- Nuclear Proteins/metabolism
- Oligonucleotides, Antisense/pharmacology
- Oxygen/metabolism
- Prostaglandin-Endoperoxide Synthases/biosynthesis
- Prostaglandins/metabolism
- RNA, Messenger/metabolism
- Recombinant Proteins/chemistry
- Sulfones
- Time Factors
- Transcription Factors
- Transcription, Genetic
- Transcriptional Activation
- Umbilical Veins/cytology
- Up-Regulation
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Paola Pichiule
- Department of Anatomy, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
104
|
Weinmann M, Marini P, Jendrossek V, Betsch A, Goecke B, Budach W, Belka C. Influence of hypoxia on TRAIL-induced apoptosis in tumor cells. Int J Radiat Oncol Biol Phys 2004; 58:386-96. [PMID: 14751507 DOI: 10.1016/j.ijrobp.2003.09.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Tumor hypoxia reduces the efficacy of radiotherapy, many types of chemotherapy, and tumor necrosis factor-alpha (TNF-alpha). TRAIL (TNF-alpha-related apoptosis-inducing ligand) is a ligand for death receptors of the TNF superfamily shown to be selectively toxic for tumor cells and thereby a promising antineoplastic tool. The impact of hypoxia on TRAIL-induced apoptosis was examined in this study. METHODS AND MATERIALS Apoptosis induction and growth rates of various tumor cell lines under hypoxia were evaluated in vitro. Biologically effective induction of hypoxia was verified by determination of hypoxia-inducible factor-1 (HIF-1) activation. The efficacy of TRAIL- and radiation-induced apoptosis under different oxygen conditions was quantified in vitro. The impact of Bcl-2 on TRAIL-induced apoptosis under hypoxia or normoxia was evaluated by comparing cells expressing Bcl-2 with a vector control. RESULTS Moderate hypoxia caused no growth retardation or apoptosis, but led to activation of HIF-1 as a prerequisite of hypoxic gene induction. Cellular responses to TRAIL differed considerably among the cell lines tested. Hypoxia reduced radiation-induced, but not TRAIL-induced, apoptosis in the tested cell lines. Hypoxia did not induce Bcl-2 expression. Bcl-2 had a minor impact on the efficacy of TRAIL-induced apoptosis. CONCLUSION Taken together, the data indicate that TRAIL is clearly effective under conditions of proven hypoxia.
Collapse
Affiliation(s)
- Martin Weinmann
- Department of Radiation Oncology, University of Tübingen, Hoppe-Seylerstrasse 3, 72076 Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
105
|
Wei Q, Alam MM, Wang MH, Yu F, Dong Z. Bid activation in kidney cells following ATP depletion in vitro and ischemia in vivo. Am J Physiol Renal Physiol 2003; 286:F803-9. [PMID: 14678945 DOI: 10.1152/ajprenal.00093.2003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Bid is a proapoptotic Bcl-2 family protein, which on activation translocates to mitochondria and induces damage to the organelles. Activation of Bid depends on its proteolytic processing into truncated forms of tBid. Bid is highly expressed in the kidneys; however, little is known about its role in renal pathophysiology. In this study, we initially examined Bid activation in cultured rat kidney proximal tubular cells following ATP depletion. The cells were depleted of ATP by azide incubation in the absence of metabolic substrates and then returned to normal culture medium for recovery. Typical apoptosis developed during recovery of ATP-depleted cells. This was accompanied by Bid cleavage, releasing tBid of 15 and 13 kDa. Bid cleavage was abolished in cells overexpressing Bcl-2, an antiapoptotic gene. It was also suppressed by caspase inhibitors. Peptide inhibitors of caspase-9 were more effective in blocking Bid cleavage compared with inhibitors of caspase-8 and caspase-3. Provision of glucose, a glycolytic substrate, during azide incubation inhibited Bid cleavage as well, indicating that Bid cleavage was initiated by ATP depletion. Consistently, Bid cleavage was also induced following ATP depletion by hypoxia or mitochondrial uncoupling. Of significance, cleaved Bid translocated to mitochondria, suggesting a role for Bid in the development of mitochondrial defects in ATP-depleted cells. Finally, Bid cleavage was induced during renal ischemia-reperfusion in the rat. Together, these results provide the first evidence for Bid activation in kidney cells following ATP depletion in vitro and renal ischemia in vivo.
Collapse
Affiliation(s)
- Qingqing Wei
- Dept. of Cellular Biology and Anatomy, Medical College of Georgia, 1459 Laney Walke Blvd., Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
106
|
Abstract
Under hypoxia, some cells are irreversibly injured and die, whereas others can adapt to the stress and survive. The molecular and genetic basis underlying cellular sensitivity to hypoxic injury is unclear. Here we have selected death-resistant cells by repeated episodes of hypoxia. The selected cells are cross-resistant to apoptosis induced by staurosporine, azide, and cisplatin. These cells up-regulate Bcl-X(L), an anti-apoptotic protein. Bcl-X(L) interacts with the pro-apoptotic molecule Bax and abrogates its toxicity in mitochondria, resulting in the preservation of mitochondrial integrity, cytochrome c, and cell viability. Down-regulation of Bcl-X(L) by antisense oligonucleotides or the newly identified Bcl-X(L) inhibitor chelerythrine restores cellular sensitivity to injury and death. Thus, Bcl-X(L) is a key molecule for hypoxia selection of death resistance. These findings may have important implications for the development of solid tumors where hypoxia selects for death-resistant cells that are inert to cancer therapy.
Collapse
Affiliation(s)
- Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | |
Collapse
|
107
|
Lenburg ME, Liou LS, Gerry NP, Frampton GM, Cohen HT, Christman MF. Previously unidentified changes in renal cell carcinoma gene expression identified by parametric analysis of microarray data. BMC Cancer 2003; 3:31. [PMID: 14641932 PMCID: PMC317310 DOI: 10.1186/1471-2407-3-31] [Citation(s) in RCA: 210] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Accepted: 11/27/2003] [Indexed: 12/15/2022] Open
Abstract
Background Renal cell carcinoma is a common malignancy that often presents as a metastatic-disease for which there are no effective treatments. To gain insights into the mechanism of renal cell carcinogenesis, a number of genome-wide expression profiling studies have been performed. Surprisingly, there is very poor agreement among these studies as to which genes are differentially regulated. To better understand this lack of agreement we profiled renal cell tumor gene expression using genome-wide microarrays (45,000 probe sets) and compare our analysis to previous microarray studies. Methods We hybridized total RNA isolated from renal cell tumors and adjacent normal tissue to Affymetrix U133A and U133B arrays. We removed samples with technical defects and removed probesets that failed to exhibit sequence-specific hybridization in any of the samples. We detected differential gene expression in the resulting dataset with parametric methods and identified keywords that are overrepresented in the differentially expressed genes with the Fisher-exact test. Results We identify 1,234 genes that are more than three-fold changed in renal tumors by t-test, 800 of which have not been previously reported to be altered in renal cell tumors. Of the only 37 genes that have been identified as being differentially expressed in three or more of five previous microarray studies of renal tumor gene expression, our analysis finds 33 of these genes (89%). A key to the sensitivity and power of our analysis is filtering out defective samples and genes that are not reliably detected. Conclusions The widespread use of sample-wise voting schemes for detecting differential expression that do not control for false positives likely account for the poor overlap among previous studies. Among the many genes we identified using parametric methods that were not previously reported as being differentially expressed in renal cell tumors are several oncogenes and tumor suppressor genes that likely play important roles in renal cell carcinogenesis. This highlights the need for rigorous statistical approaches in microarray studies.
Collapse
Affiliation(s)
- Marc E Lenburg
- Departments of Genetics & Genomics, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| | - Louis S Liou
- Urology, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| | - Norman P Gerry
- Departments of Genetics & Genomics, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| | - Garrett M Frampton
- Departments of Genetics & Genomics, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| | - Herbert T Cohen
- Medicine, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| | - Michael F Christman
- Departments of Genetics & Genomics, Boston University School of Medicine 715 Albany Street, E613 Boston, Massachusetts 02118, USA
| |
Collapse
|
108
|
Ameri K, Lewis CE, Raida M, Sowter H, Hai T, Harris AL. Anoxic induction of ATF-4 through HIF-1-independent pathways of protein stabilization in human cancer cells. Blood 2003; 103:1876-82. [PMID: 14604972 DOI: 10.1182/blood-2003-06-1859] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Hypoxia is a key factor in tumor development, contributing to angiogenesis and radiotherapy resistance. Hypoxia-inducible factor-1 (HIF-1) is a major transcription factor regulating the response of cancer cells to hypoxia. However, tumors also contain areas of more severe oxygen depletion, or anoxia. Mechanisms for survival under anoxia are HIF-1alpha independent in Caenorhabditis elegans and, thus, differ from the hypoxic response. Here we report a differential response of cancer cells to hypoxia and anoxia by demonstrating the induction of activating transcription factor-4 (ATF-4) and growth arrest DNA damage 153 (GADD153) protein specifically in anoxia and the lack of induction in hypoxia. By applying RNAi, ATF-4 induction in anoxia was shown to be independent of HIF-1alpha, and desferrioxamine mesylate (DFO) and cobalt chloride induced HIF-1alpha but not ATF-4 or GADD153. Furthermore, the inductive response of ATF-4 and GADD153 was not related to alterations in or arrest of mitochondrial respiration and was independent of von Hippel-Lindau (VHL) disease mutations. In reoxygenated anoxic cells, ATF-4 had a half-life of less than 5 minutes; adding the proteasome inhibitor to normoxic cells up-regulated ATF-4 protein. Extracts from primary human tumors demonstrated more ATF-4 expression in tumors near necrotic areas. Thus, this study demonstrates a novel HIF-1alpha-independent anoxic mechanism that regulates ATF-4 induction at the protein stability level in tumor cells.
Collapse
Affiliation(s)
- Kurosh Ameri
- Cancer Research UK, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
109
|
Bando H, Toi M, Kitada K, Koike M. Genes commonly upregulated by hypoxia in human breast cancer cells MCF-7 and MDA-MB-231. Biomed Pharmacother 2003; 57:333-40. [PMID: 14568227 DOI: 10.1016/s0753-3322(03)00098-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hypoxia is a stress that causes alterations in signal transduction and gene instability. In the cancer microenvironment, hypoxia plays a significant role in forming a tumor phenotype and tumor progression. We aimed to identify the genes upregulated by hypoxia in human breast cancer cell lines, a hormone-dependent MCF-7 and a hormone-independent MDA-MB-231, using microarray analysis. These cells were exposed to two oxygen concentrations such as 21% and 1% in a time-course. Out of 12625 genes, 26 genes were identified as commonly upregulated in both MCF-7 and MDA-MB-231 cells. Some of these genes were already reported as hypoxia-related, but some of those were identified newly. These commonly upregulated genes between hormone-dependent and hormone-independent cells would be a clue to study hypoxia-related events and to explore the novel therapeutic targets in human breast cancer.
Collapse
Affiliation(s)
- Hiroko Bando
- Breast Cancer Research Group, Tokyo Metropolitan Cancer and Infectious Diseases Center, Honkomagome, 3-18-22, Bunkyo-ku, Tokyo 113-0087, Japan
| | | | | | | |
Collapse
|
110
|
Gross J, Rheinländer C, Fuchs J, Mazurek B, Machulik A, Andreeva N, Kietzmann T. Expression of hypoxia-inducible factor-1 in the cochlea of newborn rats. Hear Res 2003; 183:73-83. [PMID: 13679140 DOI: 10.1016/s0378-5955(03)00222-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia/ischemia is a major pathogenetic factor in the development of hearing loss. An important transcription factor involved in the signaling and adaptation to hypoxia/ischemia is the hypoxia-inducible factor-1 (HIF-1). To study HIF-1 expression we used an in vitro hypoxia model of explant and dissociated cultures of the stria vascularis, the organ of Corti with limbus and the modiolus from the cochlea of 3-5-day-old Wistar rats. Hypoxia differentially increased HIF-1 activity as measured by a reporter gene. Twenty-four hour hypoxia increased HIF-1 activity 14.1+/-3.5-fold in the modiolus, 9.4+/-3.0-fold in the organ of Corti with limbus, and 6.4+/-1.5-fold in the stria vascularis. The HIF-1alpha mRNA level was measured by quantitative reverse transcription polymerase chain reaction and showed a lower expression in the modiolus (1.3+/-0.2 pg/microg RNA) than in both the organ of Corti with limbus and the stria vascularis (2.7-3.2+/-1.3, P<0.01). Hypoxia had no effect on the HIF-1alpha mRNA levels. The region-specific regulation of HIF-1 expression on the transcriptional and posttranslational levels may expand the possibilities for adaptation of the cochlea to hypoxia.
Collapse
Affiliation(s)
- Johann Gross
- Department of Otorhinolaryngology, University Hospital Charité, Spandauer Damm 130, Bldg 31, 14050 Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
111
|
Dong Z, Wang JZ, Yu F, Venkatachalam MA. Apoptosis-resistance of hypoxic cells: multiple factors involved and a role for IAP-2. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:663-71. [PMID: 12875985 PMCID: PMC1868200 DOI: 10.1016/s0002-9440(10)63693-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hypoxia is an important pathogenic factor in ischemic disease and tumorigenesis. Under hypoxia, some cells are irreversibly damaged, whereas others adapt to the stress and may become more resistant to injury. The mechanism underlying such adaptive responses is unclear. Our recent study showed hypoxic induction of inhibitor of apoptosis protein-2 (IAP-2). Here we have investigated the critical steps in the apoptotic cascade that are affected by hypoxia and have identified a role for IAP-2 in apoptosis resistance of hypoxic cells. The results show that cells cultured in hypoxia became resistant to staurosporine-induced apoptosis. Apoptosis resistance of these cells took place at the mitochondria and in the cytosol. At the mitochondrial level, membrane accumulation of the proapoptotic molecule Bax was suppressed. This was accompanied by less cytochrome c (cyt. c) release from the organelles. In the cytosol, hypoxia induced IAP-2; the cytosol with IAP-2 was resistant to cyt. c-stimulated caspase activation. Of significance, immunodepletion of IAP-2 from the hypoxic cytosol restored its competence for caspase activation. Thus, death resistance of hypoxic cells involves multiple factors targeting different stages of apoptosis, with IAP-2 suppressing caspases in the cytosol.
Collapse
Affiliation(s)
- Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | | | |
Collapse
|
112
|
Steinbach JP, Wolburg H, Klumpp A, Probst H, Weller M. Hypoxia-induced cell death in human malignant glioma cells: energy deprivation promotes decoupling of mitochondrial cytochrome c release from caspase processing and necrotic cell death. Cell Death Differ 2003; 10:823-32. [PMID: 12815465 DOI: 10.1038/sj.cdd.4401252] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia induces apoptosis in primary and transformed cells and in various tumor cell lines in vitro. In contrast, there is little apoptosis and predominant necrosis despite extensive hypoxia in human glioblastomas in vivo. We here characterize ultrastructural and biochemical features of cell death in LN-229, LN-18 and U87MG malignant glioma cells in a paradigm of hypoxia with partial glucose deprivation in vitro. Electron microscopic analysis of hypoxia-challenged glioma cells demonstrated early stages of apoptosis but predominant necrosis. ATP levels declined during hypoxia, but recovered with re-exposure to normoxic conditions unless hypoxia exceeded 8 h. Longer hypoxic exposure resulted in irreversible ATP depletion and delayed cell death. Hypoxia induced mitochondrial release of cytochrome c, but there was no cleavage of caspases 3, 7, 8 or 9, and no DNA fragmentation. Ectopic expression of BCL-XL conferred protection from hypoxia-induced cell death, whereas the overexpression of the antiapoptotic proteins X-linked-inhibitor-of-apoptosis-protein and cytokine response modifier-A had no effect. These findings suggest that glioma cells resist adverse effects of hypoxia until energy stores are depleted and then undergo necrosis rather than apoptosis because of energy deprivation.
Collapse
Affiliation(s)
- J P Steinbach
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University of Tübingen, School of Medicine, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
113
|
Wagner KD, Wagner N, Wellmann S, Schley G, Bondke A, Theres H, Scholz H. Oxygen-regulated expression of the Wilms' tumor suppressor Wt1 involves hypoxia-inducible factor-1 (HIF-1). FASEB J 2003; 17:1364-6. [PMID: 12738801 DOI: 10.1096/fj.02-1065fje] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The Wilms' tumor gene Wt1 is unique among tumor suppressors because of its requirement for the development of certain organs. We recently described de novo expression of Wt1 in myocardial blood vessels of ischemic rat hearts. The purpose of this study was to analyze the mechanism(s) of hypoxic/ischemic induction of Wt1. We show here that Wt1 mRNA and protein is up-regulated in the heart and kidneys of rats exposed to normobaric hypoxia (8% O2). Ectopic Wt1 immunoreactivity was detected in renal tubules of hypoxic rats, which also expressed the antiapoptotic protein Bcl-2 and contained significantly fewer TUNEL-positive cells than in normoxic kidneys. Wt1 expression was enhanced in the osteosarcoma line U-2OS and in Reh lymphoblast cells that were grown either at 1% O2 or in the presence of CoCl2 and desferrioxamine, respectively. The promoter of the Wt1 gene was capable of mediating expression of a luciferase reporter in response to hypoxia. We identified a hypoxia-responsive element in the Wt1 sequence that bound to hypoxia-inducible factor-1 (HIF-1) and was required for activation of the Wt1 promoter by CoCl2 and HIF-1. These findings demonstrate that Wt1 expression can be stimulated by hypoxia, which involves activation of the Wt1 promoter by HIF-1.
Collapse
|
114
|
Yi X, Yin XM, Dong Z. Inhibition of Bid-induced apoptosis by Bcl-2. tBid insertion, Bax translocation, and Bax/Bak oligomerization suppressed. J Biol Chem 2003; 278:16992-9. [PMID: 12624108 DOI: 10.1074/jbc.m300039200] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bcl-2 family proteins are important regulators of apoptosis. They can be pro-apoptotic (e.g. Bid, Bax, and Bak) or anti-apoptotic (e.g. Bcl-2 and Bcl-x(L)). The current study examined Bid-induced apoptosis and its inhibition by Bcl-2. Transfection of Bid led to apoptosis in HeLa cells. In these cells, Bid was processed into active forms of truncated Bid or tBid. Following processing, tBid translocated to the membrane-bound organellar fraction. Bcl-2 co-transfection inhibited Bid-induced apoptosis but did not prevent Bid processing or tBid translocation. On the other hand, Bcl-2 blocked the release of mitochondrial cytochrome c in Bid-transfected cells, suggesting actions at the mitochondrial level. Alkaline treatment stripped off tBid from the membrane-bound organellar fraction of Bid plus Bcl-2-co-transfected cells, but not from cells transfected with only Bid, suggesting inhibition of tBid insertion into mitochondrial membranes by Bcl-2. Bcl-2 also prevented Bid-induced Bax translocation from cytosol to the membrane-bound organellar fraction. Finally, Bcl-2 diminished Bid-induced oligomerization of Bax and Bak within the membrane-bound organellar fraction, shown by cross-linking experiments. In conclusion, Bcl-2 inhibited Bid-induced apoptosis at the mitochondrial level by blocking cytochrome c release, without suppressing Bid processing or activation. Critical steps blocked by Bcl-2 included tBid insertion, Bax translocation, and Bax/Bak oligomerization in the mitochondrial membranes.
Collapse
Affiliation(s)
- Xiaolan Yi
- Department of Anatomy and Cell Biology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | |
Collapse
|
115
|
Abstract
The crucial role of cell death in many diseases is obvious and has spurred intense research to understand the regulation of apoptotic pathways. Caspase activation is central to many of the apoptotic pathways. In recent years, the study of the regulation of caspase activation and activity in various cell lines and in diseases has revealed highly complex mechanisms regulating cell survival or cell death. In this review, the major natural cellular anticaspase factors are described with particular attention to the inhibitors that prevent active caspases from committing the cell to irreversible destruction. The major group of caspase inhibitors known is the inhibitor of apoptosis proteins (IAP) and this review describes the characteristics of IAP, regulation of IAP expression, and mechanisms of action of IAP. However, other proteins including Bcl-2 family members, heat shock proteins, caspase-like decoy, calpains and proteases, and lipid moieties in the form of phosphoinositides also can function as caspase inhibitors. The current knowledge of the inhibition of these non-IAP factors is described herein.
Collapse
Affiliation(s)
- Andréa C LeBlanc
- Department of Neurology, The Bloomfield Center for Research in Aging, H3T 1E2, Montreal, Quebec, Canada.
| |
Collapse
|
116
|
Dai S, Huang ML, Hsu CY, Chao KSC. Inhibition of hypoxia inducible factor 1alpha causes oxygen-independent cytotoxicity and induces p53 independent apoptosis in glioblastoma cells. Int J Radiat Oncol Biol Phys 2003; 55:1027-36. [PMID: 12605983 DOI: 10.1016/s0360-3016(02)04507-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE Hypoxia, which activates the hypoxia inducible factor-1 alpha (HIF-1alpha) pathway, is a common feature in malignant gliomas and has been linked with tumor cell survival and therapy resistance. In this study, we examined the effect of antisense inhibition of HIF-1alpha on the survival, apoptosis and responses to chemotherapy in U-87 malignant glioma cells. MATERIALS AND METHODS Hypoxia (1% oxygen) was achieved in a tri-gas incubator with intermittent N(2) gas flushing or in a gas tight-module sealed with 94% N(2), 1% O(2) and balance CO(2). HIF-1alpha inhibition was achieved with antisense phosphorothioate oligodeoxynucleotide (AS-HIF ODN), delivered using cytofectin GSV3815. HIF-1alpha expression level was monitored by a hypoxia-responsive luciferase reporter assay and verified by northern blot and immunoblot analyses. Cell viability was quantified by a colorimetric microtiter plate MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt] assay. Apoptotic cell death was detected with a colorimetric caspase-3 assay, as well as using terminal transferase-catalyzed in situ end labeling (TUNEL) staining. RESULTS Antisense HIF-1alpha phosphorothioate oligodeoxynucleotide (AS-HIF ODN) treatment suppressed HIF-1alpha expression by up to 80% under both normoxic and hypoxic conditions as measured by a hypoxia-responsive reporter assay and confirmed by northern and western blot analyses. Antisense knockdown of HIF-1alpha resulted in significant reduction in U-87 cells survival and an acceleration of apoptosis, which did not involve p53 transactivation. Pretreatment of cells with Z-Val-Ala-Asp (-OCH(3))-fluoromethylketone (Z-VAD), a broad-spectrum caspase inhibitor largely eliminated this effect of AS-HIF. Caspase-3 specific activity was markedly induced 3 days after AS-HIF treatment when increased cell death was also noted. Transient overexpression of HIF-1alpha in U-87 cells neutralized apoptosis-inducing effect of AS-HIF. AS-HIF treatment did not affect viability of primary astrocytes and was selectively more toxic to U-87 glioma cells than normal human fibroblasts. The HIF-1alpha antisense treatment exerted an oxygen-independent, and additive but not synergistic effect to the cytotoxicity of cisplatin, etoposide, and vincristine. CONCLUSIONS These results together indicate that suppression of HIF-1alpha-expression may be a promising strategy that is selective for reducing the survival and facilitating chemotherapeutic efficacy of malignant glioma.
Collapse
Affiliation(s)
- Simon Dai
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 77030, USA
| | | | | | | |
Collapse
|
117
|
Garayoa M, Man YG, Martínez A, Cuttitta F, Mulshine JL. Downregulation of hnRNP A2/B1 expression in tumor cells under prolonged hypoxia. Am J Respir Cell Mol Biol 2003; 28:80-5. [PMID: 12495935 DOI: 10.1165/rcmb.4880] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 has been previously shown to be overexpressed in breast and lung tumors. Because hypoxia is a feature inherent in solid tumors, the regulation of hnRNP A2/B1 expression and subcellular localization under hypoxic conditions was studied on human lung and breast carcinoma cell lines. We found that sustained hypoxic treatment downregulated hnRNP A2/B1 expression in MCF7 and H157 cell lines. Northern blot analysis showed that this decay: (i) was observed as a marked diminution of transcript levels after 24-48 h of exposure to low oxygen tension; (ii) is not mediated by the transcription factor, hypoxia inducible factor-1; and (iii) is partially dependent on a higher hnRNP A2/B1 messenger RNA turnover under hypoxic than normoxic conditions. Immunocytochemical staining also showed a significant diminution of hnRNP A2/B1 staining in these cell lines after 24-48 h of hypoxia, together with a predominant loss of cytoplasmic staining. Further investigations are warranted to evaluate the relevance of modulation of hnRNP A2/B1 in hypoxic environments relative to its previously reported utility as a marker of early lung carcinogenesis.
Collapse
Affiliation(s)
- Mercedes Garayoa
- Cell and Cancer Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
118
|
Hofer T, Wenger RH, Kramer MF, Ferreira GC, Gassmann M. Hypoxic up-regulation of erythroid 5-aminolevulinate synthase. Blood 2003; 101:348-50. [PMID: 12393745 DOI: 10.1182/blood-2002-03-0773] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The erythroid-specific isoform of 5-aminolevulinate synthase (ALAS2) catalyzes the rate-limiting step in heme biosynthesis. The hypoxia-inducible factor-1 (HIF-1) transcriptionally up-regulates erythropoietin, transferrin, and transferrin receptor, leading to increased erythropoiesis and hematopoietic iron supply. To test the hypothesis that ALAS2 expression might be regulated by a similar mechanism, we exposed murine erythroleukemia cells to hypoxia (1% O(2)) and found an up to 3-fold up-regulation of ALAS2 mRNA levels and an increase in cellular heme content. A fragment of the ALAS2 promoter ranging from -716 to +1 conveyed hypoxia responsiveness to a heterologous luciferase reporter gene construct in transiently transfected HeLa cells. In contrast, iron depletion, known to induce HIF-1 activity but inhibit ALAS2 translation, did not increase ALAS2 promoter activity. Mutation of a previously predicted HIF-1-binding site (-323/-318) within this promoter fragment and DNA-binding assays revealed that hypoxic up-regulation is independent of this putative HIF-1 DNA-binding site.
Collapse
Affiliation(s)
- Thomas Hofer
- Institute of Veterinary Physiology, University of Zurich, Switzerland
| | | | | | | | | |
Collapse
|
119
|
Budanov AV, Shoshani T, Faerman A, Zelin E, Kamer I, Kalinski H, Gorodin S, Fishman A, Chajut A, Einat P, Skaliter R, Gudkov AV, Chumakov PM, Feinstein E. Identification of a novel stress-responsive gene Hi95 involved in regulation of cell viability. Oncogene 2002; 21:6017-31. [PMID: 12203114 DOI: 10.1038/sj.onc.1205877] [Citation(s) in RCA: 298] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2002] [Revised: 07/10/2002] [Accepted: 07/15/2002] [Indexed: 01/21/2023]
Abstract
cDNA microarray hybridization was used in an attempt to identify novel genes participating in cellular responses to prolonged hypoxia. One of the identified novel genes, designated Hi95 shared significant homology to a p53-regulated GADD family member PA26. In addition to its induction in response to prolonged hypoxia, the increased Hi95 transcription was observed following DNA damage or oxidative stress, but not following hyperthermia or serum starvation. Whereas induction of Hi95 by prolonged hypoxia or by oxidative stress is most likely p53-independent, its induction in response to DNA damaging treatments (gamma- or UV-irradiation, or doxorubicin) occurs in a p53-dependent manner. Overexpression of Hi95 full-length cDNA was found toxic for many types of cultured cells directly leading either to their apoptotic death or to sensitization to serum starvation and DNA damaging treatments. Unexpectedly, conditional overexpression of the Hi95 cDNA in MCF7-tet-off cells resulted in their protection against cell death induced by hypoxia/glucose deprivation or H(2)O(2). Thus, Hi95 gene seems to be involved in complex regulation of cell viability in response to different stress conditions.
Collapse
Affiliation(s)
- Andrei V Budanov
- Department of Molecular Biology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Ave, Ohio, OH 44195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Abstract
Hypoxia-inducible factor-1 (HIF-1) is the major transcription factor specifically activated by hypoxia. It induces the expression of different genes whose products play an adaptive role for hypoxic cells and tissues. Besides these protective responses, HIF-1 and/or hypoxia have also been shown to be either anti-apoptotic or pro-apoptotic, according to the cell type and experimental conditions. More severe or prolonged hypoxia rather induces apoptosis that is, at least in part, initiated by the direct association of HIF-1alpha and p53 and p53-induced gene expression. On the other hand, HIF-1alpha dimerized with ARNT, as an active transcription factor, can protect cells from apoptosis induced by several conditions. This review is aimed to describe the different mechanisms that account for these opposite effects of HIF-1alpha.
Collapse
Affiliation(s)
- Jean-Pascal Piret
- Laboratoire de Biochimie et Biologie cellulaire, University of Namur, 61 rue de Bruxelles, 5000, Namur, Belgium
| | | | | | | |
Collapse
|
121
|
Li F, Mao HP, Ruchalski KL, Wang YH, Choy W, Schwartz JH, Borkan SC. Heat stress prevents mitochondrial injury in ATP-depleted renal epithelial cells. Am J Physiol Cell Physiol 2002; 283:C917-26. [PMID: 12176748 DOI: 10.1152/ajpcell.00517.2001] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The events that precipitate cell death and the stress proteins responsible for cytoprotection during ATP depletion remain elusive. We hypothesize that exposure to metabolic inhibitors damages mitochondria, allowing proapoptotic proteins to leak into the cytosol, and suggest that heat stress-induced hsp72 accumulation prevents mitochondrial membrane injury. To test these hypotheses, renal epithelial cells were transiently ATP depleted with sodium cyanide and 2-deoxy-D-glucose in the absence of medium dextrose. Recovery from ATP depletion was associated with the release into the cytosol of cytochrome c and apoptosis-inducing factor (AIF), proapoptotic proteins that localize to the intermitochondrial membrane space. Concomitant with mitochondrial cytochrome c leak, a seven- to eightfold increase in caspase 3 activity was observed. In controls, state III mitochondrial respiration was reduced by 30% after transient exposure to metabolic inhibitors. Prior heat stress preserved mitochondrial ATP production and significantly reduced both cytochrome c release and caspase 3 activation. Despite less cytochrome c release, prior heat stress increased binding between cytochrome c and hsp72. The present study demonstrates that mitochondrial injury accompanies exposure to metabolic inhibitors. By reducing outer mitochondrial membrane injury and by complexing with cytochrome c, hsp72 could inhibit caspase activation and subsequent apoptosis.
Collapse
Affiliation(s)
- F Li
- Renal Section, Department of Medicine, Boston Medical Center, Boston University, Massachusetts 02118-2518, USA
| | | | | | | | | | | | | |
Collapse
|
122
|
Dong Z, Nishiyama J, Yi X, Venkatachalam MA, Denton M, Gu S, Li S, Qiang M. Gene promoter of apoptosis inhibitory protein IAP2: identification of enhancer elements and activation by severe hypoxia. Biochem J 2002; 364:413-21. [PMID: 12023884 PMCID: PMC1222586 DOI: 10.1042/bj20011431] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Inhibitors of apoptosis (IAPs) antagonize cell death and regulate the cell cycle. One mechanism controlling IAP expression is translation initiation through the internal ribosome entry sites. Alternatively, IAP expression can be regulated at the transcription level. We showed recently the activation of IAP2 transcription by severe hypoxia. To pursue this regulation, we have cloned the full-length cDNA of rat IAP2, and have isolated and analysed the promoter regions of this gene. The cDNA encodes a protein of 589 amino acids, exhibiting structural features of IAP. In rat tissues, a major IAP2 transcript of approximately 3.5 kb was detected. We subsequently isolated 3.3 kb of the proximal 5'-flanking regions of this gene, which showed significant promoter activity. Of interest, 5' sequential deletion of the promoter sequence identified an enhancer of approximately 200 bp. Deletion of cAMP-response-element-binding protein (CREB) sites in the enhancer sequence diminished its activity. Finally, the IAP2 gene promoter was activated significantly by severe hypoxia and not by CoCl(2) or desferrioxamine, pharmacological inducers of hypoxia-inducible factor-1. In conclusion, in this study we have cloned the full-length cDNA of rat IAP2, and for the first time we have isolated and analysed promoter sequences of this gene, leading to the identification of enhancer elements. Moreover, we have demonstrated activation of the gene promoter by severe hypoxia, a condition shown to induce IAP2. These findings provide a basis for further investigation of gene regulation of IAP2, a protein with multiple functions.
Collapse
Affiliation(s)
- Zheng Dong
- Department of Pathology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Manderscheid M, Pereda-Fernández C, Pfeilschifter J. Cyclic AMP increases rat inhibitor of apoptosis protein (RIAP1) mRNA in renal mesangial cells. Kidney Int 2002; 61:797-803. [PMID: 11849428 DOI: 10.1046/j.1523-1755.2002.00223.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The cyclic adenosine 3',5'-monophosphate (cAMP) pathway plays a central role in the regulation of cell proliferation, differentiation and apoptosis. Cyclic AMP has been identified as a bifunctional regulator of apoptosis. The inhibitor of apoptosis proteins (IAP) regulates apoptosis by directly inhibiting distinct caspases. METHODS Expression levels of rat IAP (RIAP)-1 were investigated by RNase protection assay in rat mesangial cells after stimulation with diverse agents that modulate cellular levels of cAMP. RESULTS Rat mesangial cells up-regulated RIAP1 mRNA levels after cAMP stimulation. Membrane-permeable cAMP analogs, as well as cAMP production in response to the beta-adrenergic receptor agonist salbutamol caused a large increase in RIAP1 mRNA level, which could be inhibited by the protein kinase A inhibitors H89 and Rp-cAMPS, or by the nuclear factor-kappaB (NF-kappaB) inhibitor BAY117085. Inhibition of phosphodiesterase type IV by denbufyllin or rolipram potentiated the cAMP-mediated increase in RIAP1 mRNA. In contrast, the cyclic guanosine 3',5'-monophosphate (cGMP) analog Bt2cGMP did not affect the RIAP1 mRNA level. CONCLUSIONS These data establish, to our knowledge for the first time, that RIAP1 mRNA levels are regulated by the cAMP-signaling pathway and suggest potential new avenues of therapy to modulate apoptosis.
Collapse
Affiliation(s)
- Markus Manderscheid
- Pharmazentrum Frankfurt, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | | | |
Collapse
|
124
|
Park SY, Billiar TR, Seol DW. Hypoxia inhibition of apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Biochem Biophys Res Commun 2002; 291:150-3. [PMID: 11829475 DOI: 10.1006/bbrc.2002.6421] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia is a common environmental stress. Particularly, the center of rapidly growing solid tumors is easily exposed to hypoxic conditions. Thus, tumor cell response to hypoxia plays an important role in tumor progression as well as tumor therapy. However, little is known about hypoxic effect on apoptotic cell death. To examine the effects of hypoxia on TRAIL-induced apoptosis, human lung carcinoma A549 cells were exposed to hypoxia and treated with TRAIL protein. Hypoxia significantly protected A549 cells from apoptosis induced by TRAIL. Western blotting analysis demonstrated that hypoxia increased expression of antiapoptotic proteins such as Bcl-2, Bcl-XL, and IAP family members. The increase of these antiapoptotic molecules is believed to play an hypoxia-mediated protective role in TRAIL-induced apoptosis. Our findings suggest that an increase of antiapoptotic proteins induced by hypoxia may regulate the therapeutic activity of TRAIL protein in cancer therapy.
Collapse
Affiliation(s)
- Sang-Youel Park
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | |
Collapse
|
125
|
Abstract
Cells undergo a variety of biological responses when placed in hypoxic conditions, including activation of signalling pathways that regulate proliferation, angiogenesis and death. Cancer cells have adapted these pathways, allowing tumours to survive and even grow under hypoxic conditions, and tumour hypoxia is associated with poor prognosis and resistance to radiation therapy. Many elements of the hypoxia-response pathway are therefore good candidates for therapeutic targeting.
Collapse
Affiliation(s)
- Adrian L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
126
|
Nishiyama J, Yi X, Venkatachalam MA, Dong Z. cDNA cloning and promoter analysis of rat caspase-9. Biochem J 2001; 360:49-56. [PMID: 11695991 PMCID: PMC1222201 DOI: 10.1042/0264-6021:3600049] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Caspase-9 is the apex caspase of the mitochondrial pathway of apoptosis, which plays a critical role in apoptotic initiation and progression. However, gene regulation of caspase-9 is largely unknown. This is in part due to the lack of information on the gene promoter. Here we have cloned the full-length cDNA of rat caspase-9 and have isolated promoter regions of this gene. The rat caspase-9 cDNA of 2058 bp predicts a protein of 454 amino acids, which contains a caspase-recruitment domain ('CARD') at the N-terminus and enzymic domains at the C-terminus. The enzyme's active site, with a characteristic motif of QACGG, was also identified. Overall, rat and human caspase-9 have 71% identity. With the cDNA sequence, we subsequently isolated the proximal 5'-flanking regions of rat caspase-9 by the procedure of genomic walking. The 2270 bp genomic segment is 'TATA-less', but contains several GC boxes. Elements binding known transcription factors such as Sp-1, Pit-1, CCAAT-enhancer-binding protein (C/EBP), glucocorticoid receptor and hypoxia-inducible factor 1 (HIF-1) were also identified. When cloned into reporter gene vectors, the genomic segment showed significant promoter activity, indicating that the 5'-flanking regions isolated by genomic walking contain the gene promoter of rat caspase-9. Of significance is that the cloned promoter segments were activated by severe hypoxia, conditions inducing caspase-9 transcription. Thus, the genomic sequences reported here contain not only the basal promoter of rat caspase-9 but also regulatory elements responsive to pathophysiological stimuli including hypoxia.
Collapse
Affiliation(s)
- J Nishiyama
- Department of Pathology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|