151
|
Chen R, Le Rouzic E, Kearney JA, Mansky LM, Benichou S. Vpr-mediated incorporation of UNG2 into HIV-1 particles is required to modulate the virus mutation rate and for replication in macrophages. J Biol Chem 2004; 279:28419-25. [PMID: 15096517 DOI: 10.1074/jbc.m403875200] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human immunodeficiency virus type 1 is able to infect nondividing cells, such as macrophages, and the viral Vpr protein has been shown to participate in this process. Here, we investigated the impact of the recruitment into virus particles of the nuclear form of uracil DNA glycosylase (UNG2), a cellular DNA repair enzyme, on the virus mutation rate and on replication in macrophages. We demonstrate that the interaction of Vpr with UNG2 led to virion incorporation of a catalytically active enzyme that is directly involved with Vpr in modulating the virus mutation rate. The lack of UNG in virions during virus replication in primary monocyte-derived macrophages further exacerbated virus mutant frequencies to an 18-fold increase compared with the 4-fold increase measured in actively dividing cells. Because the presence of UNG is also critical for efficient infection of macrophages, these observations extend the role of Vpr to another early step of the virus life cycle, e.g. viral DNA synthesis, that is essential for replication of human immunodeficiency virus type 1 in nondividing cells.
Collapse
Affiliation(s)
- Renxiang Chen
- Ohio State University Biochemistry Graduate Program, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
152
|
Kino T, Pavlakis GN. Partner Molecules of Accessory Protein Vpr of the Human Immunodeficiency Virus Type 1. DNA Cell Biol 2004; 23:193-205. [PMID: 15142377 DOI: 10.1089/104454904773819789] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vpr (Viral protein-R) of the Human Immunodeficiency Virus type-1 is a 14-kDa virion-associated protein, conserved in HIV-1, -2 and the Simian Immunodeficiency Virus (SIV). Vpr is incorporated into the virion, travels to the nucleus, and has multiple activities including promoter activation, cell cycle arrest at the G2/M transition and apoptosis induction. Through these activities, Vpr is thought to influence not only viral replication but also numerous host cell functions. These functions may be categorized in three groups depending on the domains of Vpr that support them: (1) functions mediated by the amino terminal portion of Vpr, like virion packaging; (2) functions mediated by the carboxyl terminal portion such as cell cycle arrest; and (3) functions that depend on central alpha-helical structures such as transcriptional activation, apoptosis and subcellular shuttling. Association of these activities to specific regions of the Vpr molecule appears to correlate to the host/viral molecules that interact with corresponding portion of Vpr. They include Gag, host transcription factors/coactivators such as SP1, the glucocorticoid receptor, p300/CREB-binding protein and TFIIB, apoptotic adenine nucleotide translocator, cyclophilin A and 14-3-3 proteins. The properties of Vpr molecule has made it difficult to assess its function and determine the true cellular interactors. Further studies on Vpr function are needed to fully assess the function of this important early regulatory molecule of HIV and other lentiviruses.
Collapse
Affiliation(s)
- Tomoshige Kino
- Human Retrovirus Section, Center for Basic Research, National Cancer Institute-Frederick, Frederick, Maryland 21702-1201, USA
| | | |
Collapse
|
153
|
Muthumani K, Desai BM, Hwang DS, Choo AY, Laddy DJ, Thieu KP, Rao RG, Weiner DB. HIV-1 Vpr and Anti-Inflammatory Activity. DNA Cell Biol 2004; 23:239-47. [PMID: 15142381 DOI: 10.1089/104454904773819824] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
New and effective approaches for inflammatory diseases based on novel mechanisms of action are needed. One potential source of anti-inflammatory drugs exists among viruses. Viruses have evolved to infect, replicate within, and kill human cells through diverse mechanisms. They accomplish this fact by finding ways to out with the host's complex immune machinery. It is possible that the viral proteins and pathways involved in the downregulation of host immune function during infection can be exploited as a therapeutic in diseases that result in the overactivity of the immune system. Indeed, the human immunodeficiency virus type 1 (HIV-1) protein, Vpr, affects cells in a number of ways that may prove useful for exploitation for the treatment of inflammatory diseases. Vpr has effects on T-cell proliferation, cytokine production, chemokine production, and Nuclear Factor kappa B (NF-kappaB)-mediated transcription. Importantly, it has been observed that Vpr downregulates NF-kappaB and the production of pro-inflammatory cytokines such as TNF-alpha, and IL-12. These activities are worthy of further examination for control of hyperinflammatory and hyperproliferative conditions.
Collapse
Affiliation(s)
- Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
154
|
Griffin SDC, Harvey R, Clarke DS, Barclay WS, Harris M, Rowlands DJ. A conserved basic loop in hepatitis C virus p7 protein is required for amantadine-sensitive ion channel activity in mammalian cells but is dispensable for localization to mitochondria. J Gen Virol 2004; 85:451-461. [PMID: 14769903 DOI: 10.1099/vir.0.19634-0] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We previously identified the function of the hepatitis C virus (HCV) p7 protein as an ion channel in artificial lipid bilayers and demonstrated that this in vitro activity is inhibited by amantadine. Here we show that the ion channel activity of HCV p7 expressed in mammalian cells can substitute for that of influenza virus M2 in a cell-based assay. This was also the case for the p7 from the related virus, bovine viral diarrhoea virus (BVDV). Moreover, amantadine was shown to abrogate HCV p7 function in this assay at a concentration that specifically inhibits M2. Mutation of a conserved basic loop located between the two predicted trans-membrane alpha helices rendered HCV p7 non-functional as an ion channel. The intracellular localization of p7 was unaffected by this mutation and was found to overlap significantly with membranes associated with mitochondria. Demonstration of p7 ion channel activity in cellular membranes and its inhibition by amantadine affirm the protein as a target for future anti-viral chemotherapy.
Collapse
Affiliation(s)
- Stephen D C Griffin
- Astbury Centre of Molecular Biology, School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, UK
| | - Ruth Harvey
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Dean S Clarke
- Astbury Centre of Molecular Biology, School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, UK
| | - Wendy S Barclay
- School of Animal and Microbial Sciences, University of Reading, Whiteknights, PO Box 228, Reading, Berkshire RG6 6AJ, UK
| | - Mark Harris
- Astbury Centre of Molecular Biology, School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, UK
| | - David J Rowlands
- Astbury Centre of Molecular Biology, School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
155
|
Lecoeur H, Langonné A, Baux L, Rebouillat D, Rustin P, Prévost MC, Brenner C, Edelman L, Jacotot E. Real-time flow cytometry analysis of permeability transition in isolated mitochondria. Exp Cell Res 2004; 294:106-17. [PMID: 14980506 DOI: 10.1016/j.yexcr.2003.10.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2003] [Revised: 09/29/2003] [Indexed: 12/16/2022]
Abstract
Mitochondrial membrane permeabilization (MMP) is a key event in necrotic and (intrinsic) apoptotic processes. MMP is controlled by a few major rate-limiting events, one of which is opening of the permeability transition pore (PTP). Here we develop a flow cytometry (FC)-based approach to screen and study inducers and blockers of MMP in isolated mitochondria. Fixed-time and real-time FC permits to co-evaluate and order modifications of mitochondrial size, structure and inner membrane (IM) electrochemical potential (DeltaPsi(m)) during MMP. Calcium, a major PTP opener, and alamethicin, a PTP-independent MMP inducer, trigger significant mitochondrial forward scatter (FSC) increase and side scatter (SSC) decrease, correlating with spectrophotometrically detected swelling. FC-based fluorescence detection of the DeltaPsi(m)-sensitive cationic lipophilic dye JC-1 permits to detect DeltaPsi(m) variations induced by PTP openers or specific inducers of inner MMP such as carbonylcyanide m-chlorophenylhydrazone (mClCCP). These simple, highly sensitive and quantitative FC-based methods will be pertinent to evaluate compounds for their ability to control MMP.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Theraptosis Research Laboratory, Theraptosis S.A., Institut Pasteur, 75015 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Bruns K, Fossen T, Wray V, Henklein P, Tessmer U, Schubert U. Structural characterization of the HIV-1 Vpr N terminus: evidence of cis/trans-proline isomerism. J Biol Chem 2003; 278:43188-201. [PMID: 12881523 DOI: 10.1074/jbc.m305413200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The 96-residue human immunodeficiency virus (HIV) accessory protein Vpr serves manifold functions in the retroviral life cycle including augmentation of viral replication in non-dividing host cells, induction of G2 cell cycle arrest, and modulation of HIV-induced apoptosis. Using a combination of dynamic light scattering, circular dichroism, and NMR spectroscopy the N terminus of Vpr is shown to be a unique domain of the molecule that behaves differently from the C-terminal domain in terms of self-association and secondary structure folding. Interestingly, the four highly conserved proline residues in the N terminus are predicted to have a high propensity for cis/trans isomerism. Thus the high resolution structure and folding of a synthetic N-terminal peptide (Vpr1-40) and smaller fragments thereof have been investigated. 1H NMR data indicate Vpr1-40 possesses helical structure between residues 17-32, and for the first time, this helix, which is bound by proline residues, was observed even in aqueous solution devoid of any detergent supplements. In addition, NMR data revealed that all of the proline residues undergo a cis/ trans isomerism to such an extent that approximately 40% of all Vpr molecules possess at least one proline in a cis conformation. This phenomenon of cis/trans isomerism, which is unprecedented for HIV-1 Vpr, not only provides an explanation for the molecular heterogeneity observed in the full-length molecule but also indicates that in vivo the folding and function of Vpr should depend on a cis/trans-proline isomerase activity, particularly as two of the proline residues in positions 14 and 35 show considerable amounts of cis isomers. This prediction correlates well with our recent observation (Zander, K., Sherman, M. P., Tessmer, U., Bruns, K., Wray, V., Prechtel, A. T., Schubert, E., Henklein, P., Luban, J., Neidleman, J., Greene, W. C., and Schubert, U. (2003) J. Biol. Chem. 278, 43170-43181) of a functional interaction between the major cellular isomerase cyclophilin A and Vpr, both of which are incorporated into HIV-1 virions.
Collapse
Affiliation(s)
- Karsten Bruns
- Department of Structural Biology, Gesellschaft für Biotechnologische Forschung, D-38124 Braunschweig, Germany.
| | | | | | | | | | | |
Collapse
|
157
|
Alimonti JB, Ball TB, Fowke KR. Mechanisms of CD4+ T lymphocyte cell death in human immunodeficiency virus infection and AIDS. J Gen Virol 2003; 84:1649-1661. [PMID: 12810858 DOI: 10.1099/vir.0.19110-0] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIDS, caused by the retroviruses human immunodeficiency virus type 1 and type 2 (HIV-1 and HIV-2), has reached pandemic proportions. Therefore, it is critical to understand how HIV causes AIDS so that appropriate therapies can be formulated. Primarily, HIV infects and kills CD4(+) T lymphocytes, which function as regulators and amplifiers of the immune response. In the absence of effective anti-retroviral therapy, the hallmark decrease in CD4(+) T lymphocytes during AIDS results in a weakened immune system, impairing the body's ability to fight infections or certain cancers such that death eventually ensues. The major mechanism for CD4(+) T cell depletion is programmed cell death (apoptosis), which can be induced by HIV through multiple pathways. Death of HIV-infected cells can result from the propensity of infected lymphocytes to form short-lived syncytia or from an increased susceptibility of the cells to death. However, the apoptotic cells appear to be primarily uninfected bystander cells and are eradicated by two different mechanisms: either a Fas-mediated mechanism during activation-induced cell death (AICD), or as a result of HIV proteins (Tat, gp120, Nef, Vpu) released from infected cells stimulating apoptosis in uninfected bystander cells. There is also evidence that as AIDS progresses cytokine dysregulation occurs, and the overproduction of type-2 cytokines (IL-4, IL-10) increases susceptibility to AICD whereas type-1 cytokines (IL-12, IFN-gamma) may be protective. Clearly there are multiple causes of CD4(+) T lymphocyte apoptosis in AIDS and therapies that block or decrease that death could have significant clinical benefit.
Collapse
Affiliation(s)
- Judie B Alimonti
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 539-730 William Avenue, Winnipeg, MB, Canada R3E 0W3
| | - T Blake Ball
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 539-730 William Avenue, Winnipeg, MB, Canada R3E 0W3
| | - Keith R Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 539-730 William Avenue, Winnipeg, MB, Canada R3E 0W3
| |
Collapse
|
158
|
Tronstad KJ, Gjertsen BT, Krakstad C, Berge K, Brustugun OT, Døskeland SO, Berge RK. Mitochondrial-targeted fatty acid analog induces apoptosis with selective loss of mitochondrial glutathione in promyelocytic leukemia cells. CHEMISTRY & BIOLOGY 2003; 10:609-18. [PMID: 12890534 DOI: 10.1016/s1074-5521(03)00142-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Some fatty acids and derivatives are known to induce cell death in cancer cells. Mitochondria may have important roles in the death process. Therefore, we investigated the mitochondrial contribution in cell death induced by a modified fatty acid, tetradecylthioacetic acid (TTA), which cannot be beta-oxidized. TTA treatment induced apoptosis in IPC-81 leukemia cells via depolarization of the mitochondrial membrane potential (deltapsi) and early release of cytochrome c, accompanied by depletion of mitochondrial glutathione. Caspase-3 activation and cleavage of poly (ADP-ribose) polymerase (PARP) occurred at a late stage, but the broad-spectra caspase inhibitor zVAD-fmk did not block TTA-induced apoptosis. Overexpression of Bcl-2 partially prevented TTA-induced apoptosis, whereas cAMP-induced cell death was completely blocked. In conclusion, TTA seems to trigger apoptosis through mitochondrial-mediated mechanisms and selective modulation of the mitochondrial redox equilibrium.
Collapse
Affiliation(s)
- Karl Johan Tronstad
- Section of Medical Biochemistry, Institute of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | | | | | | | | | | | | |
Collapse
|
159
|
Badley AD, Roumier T, Lum JJ, Kroemer G. Mitochondrion-mediated apoptosis in HIV-1 infection. Trends Pharmacol Sci 2003; 24:298-305. [PMID: 12823956 DOI: 10.1016/s0165-6147(03)00125-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Acquired immunodeficiency syndrome (AIDS), which is caused by human immunodeficiency virus (HIV-1), involves the apoptotic destruction of lymphocytes and, in the context of AIDS-associated pathologies, of neurons and myocytes. Several proteins encoded by HIV-1 trigger apoptosis by inducing permeabilization of the mitochondrial membrane. Several nucleoside analogs used clinically in the treatment of HIV-1 inhibit the replication of mitochondrial DNA (mtDNA) and/or increase the frequency of mtDNA mutations. These cause severe mitochondriopathy and might contribute to lipodystrophy, the complication associated with HIV-1 therapy. HIV-1 protease inhibitors can inhibit apoptosis at the mitochondrial level, which might help to alleviate lymphopenia. Thus, it appears that the pathogenesis of AIDS, and the pharmacological interventions and complications associated with this disease, affect the mitochondrial regulation of apoptosis, which, therefore, largely determines the outcome of HIV-1 infection.
Collapse
Affiliation(s)
- Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester MN 55905, USA.
| | | | | | | |
Collapse
|
160
|
Lum JJ, Cohen OJ, Nie Z, Weaver JG, Gomez TS, Yao XJ, Lynch D, Pilon AA, Hawley N, Kim JE, Chen Z, Montpetit M, Sanchez-Dardon J, Cohen EA, Badley AD. Vpr R77Q is associated with long-term nonprogressive HIV infection and impaired induction of apoptosis. J Clin Invest 2003; 111:1547-54. [PMID: 12750404 PMCID: PMC155040 DOI: 10.1172/jci16233] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The absence of immune defects that occurs in the syndrome of long-term nonprogressive (LTNP) HIV infection offers insights into the pathophysiology of HIV-induced immune disease. The (H[F/S]RIG)(2) domain of viral protein R (Vpr) induces apoptosis and may contribute to HIV-induced T cell depletion. We demonstrate a higher frequency of R77Q Vpr mutations in patients with LTNP than in patients with progressive disease. In addition, T cell infections using vesicular stomatitis virus G (VSV-G) pseudotyped HIV-1 Vpr R77Q result in less (P = 0.01) T cell death than infections using wild-type Vpr, despite similar levels of viral replication. Wild-type Vpr-associated events, including procaspase-8 and -3 cleavage, loss of mitochondrial transmembrane potential (deltapsi(m)), and DNA fragmentation factor activation are attenuated by R77Q Vpr. These data highlight the pathophysiologic role of Vpr in HIV-induced immune disease and suggest a novel mechanism of LTNP.
Collapse
Affiliation(s)
- Julian J Lum
- Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Brenner C, Kroemer G. The mitochondriotoxic domain of Vpr determines HIV-1 virulence. J Clin Invest 2003; 111:1455-7. [PMID: 12750393 PMCID: PMC155055 DOI: 10.1172/jci18609] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Catherine Brenner
- Centre National de la Recherche Scientifique, Formation de Recherche en Evolution 2445, Université de Versailles/St. Quentin,Versailles, France
| | | |
Collapse
|
162
|
Abstract
Granzyme B is a serine proteinase that acts as a key effector of cell death mediated by cytotoxic T lymphocytes. The enzyme is transferred from the cytotoxic cell to the pathogenic target cell where it cleaves and activates a number of substrates involved in the induction of apoptosis. However, recent evidence implicates mitochondria as playing an important role in both the initiation of apoptosis and control of substrate cleavage by granzyme B in cytotoxic T lymphocyte induced death. This review focuses on current research in this rapidly expanding field, specifically the role of mitochondria in cell death induced by components of cytotoxic granules in particular granzyme B.
Collapse
Affiliation(s)
- Darren L Roberts
- Department of Biochemistry, Room 463, Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | | | |
Collapse
|
163
|
Muthumani K, Choo AY, Hwang DS, Chattergoon MA, Dayes NN, Zhang D, Lee MD, Duvvuri U, Weiner DB. Mechanism of HIV-1 viral protein R-induced apoptosis. Biochem Biophys Res Commun 2003; 304:583-92. [PMID: 12729593 DOI: 10.1016/s0006-291x(03)00631-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The paradigm of HIV-1 infection includes the diminution of CD4(+) T cells, loss of immune function, and eventual progression to AIDS. However, the mechanisms that drive host T cell depletion remain elusive. One HIV protein thought to participate in this destructive cascade is the Vpr gene product. Accordingly, we review the biology of the HIV-1 viral protein R (Vpr) an apoptogenic HIV-1 accessory protein that is packaged into the virus particle. In this review we focus specifically on Vpr's ability to induce host cell apoptosis. Recent evidence suggests that Vpr implements a unique mechanism to drive host cell apoptosis, by directly depolarizing the mitochondria membrane potential. Vpr's attack on the mitochondria results in release of cytochrome c resulting in activation of the caspase 9 pathway culminating in the activation of caspase 3 and the downstream events of apoptosis. Vpr may interact with the adenine nucleotide translocator (ANT) to prompt this cascade. The role of Vpr-induced apoptosis in HIV pathogenesis is considered.
Collapse
Affiliation(s)
- Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 505 Stellar Chance Laboratories, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Abstract
It is now well admitted that HIV infection leading to AIDS is associated with an abnormal susceptibility of T cells to undergo apoptosis. Recent progress in research into programmed cell death has resulted in the identification of the principal pathways involved in this process. Thus the "extrinsic" as well as the "intrinsic" pathways converge to the mitochondria considered as the main sensor of programmed cell death. This review summarizes our knowledge of the influence of mitochondrial control on T cell death during HIV and SIV infections.
Collapse
Affiliation(s)
- Damien Arnoult
- EMI-U 9922 INSERM/Université Paris 7, IFR02, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | | | | |
Collapse
|
165
|
Boya P, Roumier T, Andreau K, Gonzalez-Polo RA, Zamzami N, Castedo M, Kroemer G. Mitochondrion-targeted apoptosis regulators of viral origin. Biochem Biophys Res Commun 2003; 304:575-81. [PMID: 12729592 DOI: 10.1016/s0006-291x(03)00630-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During coevolution with their hosts, viruses have "learned" to intercept or to activate the principal signal transducing pathways leading to cell death. A number of proteins from pathophysiologically relevant viruses are targeted to mitochondria and regulate (induce or inhibit) the apoptosis-associated permeabilization of mitochondrial membranes. Such proteins are encoded by human immunodeficiency virus 1, Kaposi's sarcoma-associated herpesvirus, human T-cell leukemia virus-1, hepatitis B virus, cytomegalovirus, and Epstein Barr virus, among others. Within mitochondria, such apoptosis regulators from viral origin can target distinct proteins from the Bcl-2 family and the permeability transition pore complex including the adenine nucleotide translocase, cyclophilin D, the voltage-dependent anion channel, and the peripheral benzodiazepine receptor. Thus, viral proteins can regulate apoptosis at the mitochondrial level by acting on a variety of different targets.
Collapse
Affiliation(s)
- Patricia Boya
- Centre National de la Recherche Scientifique, UMR 8125, Institut Gustave Roussy, Pavillon de Recherche 1, 39 rue Camille Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
166
|
Hockenbery DM, Giedt CD, O'Neill JW, Manion MK, Banker DE. Mitochondria and apoptosis: new therapeutic targets. Adv Cancer Res 2003; 85:203-42. [PMID: 12374287 DOI: 10.1016/s0065-230x(02)85007-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- David M Hockenbery
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | | | | | |
Collapse
|
167
|
Solary E, Bettaieb A, Dubrez-Daloz L, Corcos L. Mitochondria as a target for inducing death of malignant hematopoietic cells. Leuk Lymphoma 2003; 44:563-74. [PMID: 12769332 DOI: 10.1080/1042819021000038001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Mitochondria plays a central role in apoptotic cell death. The intermembrane space of mitochondria contains a number of soluble molecules whose release from the organelle to the cytosol or the nucleus induces cell death. Thus, molecules that directly trigger mitochondria membrane permeabilisation are efficient cytotoxic drugs. Mitochondria is one of the cellular targets for commonly used epipodophyllotoxins, adenine deoxynucleoside analogs and taxanes as well as recently developped agents such as the pentacyclic triterpene betulinic acid and the lymphotoxic agent FTY720. Most informations on anthracyclines point to the mitochondrial membrane as the main target of cardiotoxicity. Mitochondria is also a target for arsenite trioxide, an old cytotoxic agent recently used for treating acute promyelocytic leukemia, lonidamine, a dichlorinated derivative of indazole-3-carboxylic acid developped as a chemosensitizer, the retinoic acid receptor gamma activator CD437 and nitric oxide (NO). Recently, cytotoxic drugs have been specifically designed to directly affect the mitochondrial function. These include the positively charged alpha-helical peptides, which are attracted to and disrupt the negatively charged mitochondrial membrane, thus inducing mammalian cell apoptosis when targeted intracellularly. Various strategies have been proposed also to directly inhibit Bcl-2 and related anti-apoptotic proteins, including antisense oligonucleotides (e.g. Genasense, currently tested in phase III trials), small molecules that mimic the BH3 dimerization domain of these proteins and kinase inhibitors. Ligands of the mitochondrial benzodiazepine receptor such as the isoquinolone carboxamide derivative PK11195 also overcome the membrane-stabilizing effect of Bcl-2, whereas the adenosine nucleotide translocator (ANT) and the mitochondrial DNA are two other potential cellular targets for cytotoxic agents. Potentially, new compounds directly targeting the mitochondria may be useful in treating hematological malignancies. The challenge is now to selectively target these mitochondria permeabilizing agents to malignant cells. This review briefly summarizes the role of the mitochondria in cell death and describes these various strategies for targeting the mitochondria to induce apoptosis.
Collapse
Affiliation(s)
- Eric Solary
- INSERM U517, IFR 100, 7 boulevard Jeanne d'Arc, 21000 Dijon, France.
| | | | | | | |
Collapse
|
168
|
Abstract
The application of surface plasmon resonance (SPR)-based optical biosensors has contributed extensively to our understanding of functional aspects of HIV. SPR biosensors allow the analysis of real-time interactions of any biomolecule, be it protein, nucleic acid, lipid, carbohydrate or small molecule, without the need for intrinsic or extrinsic probes. As such, the technology has been used to analyze molecular interactions associated with every aspect of the viral life cycle, from basic studies of binding events occurring during docking, replication, budding and maturation to applied research related to vaccine and inhibitory drug development. Along the way, SPR biosensors have provided a unique and detailed view into the inner workings of HIV.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, School of Medicine, Medical Drive, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
169
|
Nudson WA, Rovnak J, Buechner M, Quackenbush SL. Walleye dermal sarcoma virus Orf C is targeted to the mitochondria. J Gen Virol 2003; 84:375-381. [PMID: 12560570 DOI: 10.1099/vir.0.18570-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Walleye dermal sarcomas are associated with the presence of a complex retrovirus, walleye dermal sarcoma virus (WDSV). These sarcomas develop and regress seasonally in naturally infected fish. In addition to gag, pol and env, WDSV contains three open reading frames (ORFs), designated orf a, orf b and orf c. orf c is located between the 5' long terminal repeat and gag. Developing tumours contain low levels of orf a and orf b transcripts, whereas regressing tumours contain high levels of genomic transcripts and virus particles. Orf C protein is encoded by the full-length, genomic transcript and can be detected in tumour extracts with anti-Orf C-specific antisera. To determine the subcellular location of WDSV Orf C, cultured cells were transfected with an expression vector encoding haemagglutinin-tagged Orf C and examined by immunofluorescence. Orf C was observed throughout the cytoplasm and accumulated in cytoplasmic organelles. Dual-antibody staining for Orf C and mitochondrial cytochrome c demonstrated colocalization of Orf C with mitochondria and loss of the normal distribution of mitochondria in the cytoplasm. Cells transiently expressing Orf C exhibited apoptotic morphology and increased levels of surface phosphatidylserine and were unable to retain MitoTracker Orange, a dye that accumulates in active mitochondria. These results imply a functional role for WDSV Orf C in an alteration of mitochondrial function that results in apoptosis contributing to tumour regression.
Collapse
Affiliation(s)
- Wade A Nudson
- Department of Molecular Biosciences, University of Kansas, 7047 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS 66045-7534, USA
| | - Joel Rovnak
- Department of Molecular Biosciences, University of Kansas, 7047 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS 66045-7534, USA
| | - Matthew Buechner
- Department of Molecular Biosciences, University of Kansas, 7047 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS 66045-7534, USA
| | - Sandra L Quackenbush
- Department of Molecular Biosciences, University of Kansas, 7047 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS 66045-7534, USA
| |
Collapse
|
170
|
García-Ruiz C, Colell A, Marí M, Morales A, Calvo M, Enrich C, Fernández-Checa JC. Defective TNF-alpha-mediated hepatocellular apoptosis and liver damage in acidic sphingomyelinase knockout mice. J Clin Invest 2003; 111:197-208. [PMID: 12531875 PMCID: PMC151862 DOI: 10.1172/jci16010] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This study addressed the contribution of acidic sphingomyelinase (ASMase) in TNF-alpha-mediated hepatocellular apoptosis. Cultured hepatocytes depleted of mitochondrial glutathione (mGSH) became sensitive to TNF-alpha, undergoing a time-dependent apoptotic cell death preceded by mitochondrial membrane depolarization, cytochrome c release, and caspase activation. Cyclosporin A treatment rescued mGSH-depleted hepatocytes from TNF-alpha-induced cell death. In contrast, mGSH-depleted hepatocytes deficient in ASMase were resistant to TNF-alpha-mediated cell death but sensitive to exogenous ASMase. Furthermore, although in vivo administration of TNF-alpha or LPS to galactosamine-pretreated ASMase(+/+) mice caused liver damage, ASMase(-/-) mice exhibited minimal hepatocellular injury. To analyze the requirement of ASMase, we assessed the effect of glucosylceramide synthetase inhibition on TNF-alpha-mediated apoptosis. This approach, which blunted glycosphingolipid generation by TNF-alpha, protected mGSH-depleted ASMase(+/+) hepatocytes from TNF-alpha despite enhancement of TNF-alpha-stimulated ceramide formation. To further test the involvement of glycosphingolipids, we focused on ganglioside GD3 (GD3) because of its emerging role in apoptosis through interaction with mitochondria. Analysis of the cellular redistribution of GD3 by laser scanning confocal microscopy revealed the targeting of GD3 to mitochondria in ASMase(+/+) but not in ASMase(-/-) hepatocytes. However, treatment of ASMase(-/-) hepatocytes with exogenous ASMase induced the colocalization of GD3 and mitochondria. Thus, ASMase contributes to TNF-alpha-induced hepatocellular apoptosis by promoting the mitochondrial targeting of glycosphingolipids.
Collapse
Affiliation(s)
- Carmen García-Ruiz
- Liver Unit, Instituto de Malalties Digestives, Hospital Clinic i Provincial, Instituto de Investigaciones Biomédicas August Pi Suñer, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
171
|
Notario B, Zamora M, Viñas O, Mampel T. All-trans-retinoic acid binds to and inhibits adenine nucleotide translocase and induces mitochondrial permeability transition. Mol Pharmacol 2003; 63:224-31. [PMID: 12488555 DOI: 10.1124/mol.63.1.224] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the effects of retinoic acids on mitochondrial permeability transition (MPT) measured as changes in rhodamine 123 fluorescence from both isolated heart mitochondria and HeLa cells. We report that all-trans-retinoic acid (atRA), 9-cis-retinoic acid, and 13-cis-retinoic acid induce a drop in mitochondrial membrane potential in isolated mitochondria. The atRA effect was done through the induction of MPT because it was dependent on Ca(2+), in a synergic mechanism, and inhibited by cyclosporin A (CsA). Furthermore, atRA also opened MPT in vivo, because treatment of HeLa cells with atRA results in a CsA-sensitive drop of mitochondrial membrane potential. We demonstrated for the first time that retinoic acids inhibit adenine nucleotide translocase (ANT) activity in heart and liver mitochondria. Kinetic studies revealed atRA as an uncompetitive inhibitor of ANT. Photoaffinity labeling of mitochondrial proteins with [3H]atRA demonstrated the binding of a 31-kDa protein to atRA. This protein was identified as ANT because the presence of carboxyatractyloside, a specific ANT inhibitor, prevented labeling. The specific photolabeling of ANT was also prevented in a concentration-dependent manner by nonlabeled atRA, whereas palmitic acid was ineffective. This study indicates that specific interaction between atRA and ANT takes place regulating MPT opening and adenylate transport. These observations establish a novel mechanism for atRA action, which could control both energetic and apoptotic mitochondrial processes in situations such as retinoic acid treatment.
Collapse
Affiliation(s)
- Belen Notario
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain
| | | | | | | |
Collapse
|
172
|
Yao XJ, Lemay J, Rougeau N, Clément M, Kurtz S, Belhumeur P, Cohen EA. Genetic selection of peptide inhibitors of human immunodeficiency virus type 1 Vpr. J Biol Chem 2002; 277:48816-26. [PMID: 12379652 DOI: 10.1074/jbc.m207982200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) encodes a gene product, Vpr, that facilitates the nuclear uptake of the viral pre-integration complex in non-dividing cells and causes infected cells to arrest in the G(2) phase of the cell cycle. Vpr was also shown to cause mitochondrial dysfunction in human cells and budding yeasts, an effect that was proposed to lead to growth arrest and cell killing in budding yeasts and apoptosis in human cells. In this study, we used a genetic selection in Saccharomyces cerevisiae to identify hexameric peptides that suppress the growth arrest phenotype mediated by Vpr. Fifteen selected glutathione S-transferase (GST)-fused peptides were found to overcome to different extents Vpr-mediated growth arrest. Amino acid analysis of the inhibitory peptide sequences revealed the conservation of a di-tryptophan (diW) motif. DiW-containing GST-peptides interacted with Vpr in GST pull-down assays, and their level of interaction correlated with their ability to overcome Vpr-mediated growth arrest. Importantly, Vpr-binding GST-peptides were also found to alleviate Vpr-mediated apoptosis and G(2) arrest in HIV-1-producing CD4(+) T cell lines. Furthermore, they co-localized with Vpr and interfered with its nuclear translocation. Overall, this study defines a class of diW-containing peptides that inhibit HIV-1 Vpr biological activities most likely by interacting with Vpr and interfering with critical protein interactions.
Collapse
Affiliation(s)
- Xiao-Jian Yao
- Laboratoire de Rétrovirologie Humaine, Département de Microbiologie et Immunologie, Faculté de Médecine, Université de Montréal, Québec H3C 3J7, Canada
| | | | | | | | | | | | | |
Collapse
|
173
|
Debatin KM, Poncet D, Kroemer G. Chemotherapy: targeting the mitochondrial cell death pathway. Oncogene 2002; 21:8786-803. [PMID: 12483532 DOI: 10.1038/sj.onc.1206039] [Citation(s) in RCA: 303] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2002] [Revised: 09/09/2002] [Accepted: 09/11/2002] [Indexed: 12/31/2022]
Abstract
One of the mechanisms by which chemotherapeutics destroy cancer cells is by inducing apoptosis. Apoptosis can be activated through several different signalling pathways, but these all appear to converge at a single event - mitochondrial membrane permeabilization (MMP). This 'point-of-no-return' in the cell death program is a complex process that is regulated by the composition of the mitochondrial membrane and pre-mitochondrial signal-transduction events. MMP is subject to a complex regulation, and local alterations in the composition of mitochondrial membranes, as well as alterations in pre-mitochondrial signal-transducing events, can determine chemotherapy resistance in cancer cells. Detecting MMP might thus be useful for detecting chemotherapy responses in vivo. Several cytotoxic drugs induce MMP by a direct action on mitochondria. This type of agents can enforce death in cells in which upstream signals normally leading to apoptosis have been disabled. Cytotoxic components acting on mitochondria can specifically target proteins from the Bcl-2 family, the peripheral benzodiazepin receptor, or the adenine nucleotide translocase, and/or act by virtue of their physicochemical properties as steroid analogues, cationic ampholytes, redox-active compounds or photosensitizers. Some compounds acting on mitochondria can overcome the cytoprotective effect of Bcl-2-like proteins. Several agents which are already used in anti-cancer chemotherapy can induce MMP, and new drugs specifically designed to target mitochondria are being developed.
Collapse
|
174
|
Castedo M, Perfettini JL, Roumier T, Kroemer G. Cyclin-dependent kinase-1: linking apoptosis to cell cycle and mitotic catastrophe. Cell Death Differ 2002; 9:1287-93. [PMID: 12478465 DOI: 10.1038/sj.cdd.4401130] [Citation(s) in RCA: 280] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2002] [Accepted: 08/09/2002] [Indexed: 11/08/2022] Open
Abstract
The cyclin-dependent kinase 1 (Cdk1), formerly called Cdc2 (or p34(Cdc2)), interacts with cyclin B1 to form an active heterodimer. The activity of Cdk1 is subjected to a complex spatiotemporary regulation, required to guarantee its scheduled contribution to the mitotic prophase and metaphase. Moreover, the activation of Cdk1 may be required for apoptosis induction in some particular pathways of cell killing. This applies to several clinically important settings, for instance to paclitaxel-induced killing of breast cancer cells, in which the ErbB2 receptor kinase can mediate apoptosis inhibition through inactivation of Cdk1. The activation of Cdk1 participates also in HIV-1-induced apoptosis, upstream of the p53-dependent mitochondrial permeabilization step. An unscheduled Cdk1 activation may contribute to neuronal apoptosis occurring in neurodegenerative diseases. Finally, the premature activation of Cdk1 can lead to mitotic catastrophe, for instance after irradiation-induced DNA damage. Thus, a cell type-specific modulation of Cdk1 might be taken advantage of for the therapeutic correction of pathogenic imbalances in apoptosis control.
Collapse
Affiliation(s)
- M Castedo
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif, France
| | | | | | | |
Collapse
|
175
|
Everett H, Barry M, Sun X, Lee SF, Frantz C, Berthiaume LG, McFadden G, Bleackley RC. The myxoma poxvirus protein, M11L, prevents apoptosis by direct interaction with the mitochondrial permeability transition pore. J Exp Med 2002; 196:1127-39. [PMID: 12417624 PMCID: PMC2194110 DOI: 10.1084/jem.20011247] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
M11L, an antiapoptotic protein essential for the virulence of the myxoma poxvirus, is targeted to mitochondria and prevents the loss of mitochondrial membrane potential that accompanies cell death. In this study we show, using a cross-linking approach, that M11L physically associates with the mitochondrial peripheral benzodiazepine receptor (PBR) component of the permeability transition (PT) pore. Close association of M11L and the PBR is also indicated by fluorescence resonance energy transfer (FRET) analysis. Stable expression of M11L prevents the release of mitochondrial cytochrome c induced by staurosporine or protoporphyrin IX (PPIX), a ligand of the PBR. Transiently expressed M11L also prevents mitochondrial membrane potential loss induced by PPIX, or induced by staurosporine in combination with PK11195, another ligand of the PBR. Myxoma virus infection and the associated expression of early proteins, including M11L, protects cells from staurosporine- and Fas-mediated mitochondrial membrane potential loss and this effect is augmented by the presence of PBR. We conclude that M11L regulates the mitochondrial permeability transition pore complex, most likely by direct modulation of the PBR.
Collapse
Affiliation(s)
- Helen Everett
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G2H7, Canada
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Castedo M, Perfettini JL, Kroemer G. Mitochondrial apoptosis and the peripheral benzodiazepine receptor: a novel target for viral and pharmacological manipulation. J Exp Med 2002; 196:1121-5. [PMID: 12417623 PMCID: PMC2194100 DOI: 10.1084/jem.20021758] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Maria Castedo
- Centre National de la Recherche Scientifique, UMR 1599, Institut Gustave Roussy, F-94805 Villejuif, France
| | | | | |
Collapse
|
177
|
Abstract
We have assembled references of 700 articles published in 2001 that describe work performed using commercially available optical biosensors. To illustrate the technology's diversity, the citation list is divided into reviews, methods and specific applications, as well as instrument type. We noted marked improvements in the utilization of biosensors and the presentation of kinetic data over previous years. These advances reflect a maturing of the technology, which has become a standard method for characterizing biomolecular interactions.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
178
|
Roumier T, Vieira HLA, Castedo M, Ferri KF, Boya P, Andreau K, Druillennec S, Joza N, Penninger JM, Roques B, Kroemer G. The C-terminal moiety of HIV-1 Vpr induces cell death via a caspase-independent mitochondrial pathway. Cell Death Differ 2002; 9:1212-9. [PMID: 12404120 DOI: 10.1038/sj.cdd.4401089] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2002] [Revised: 05/09/2002] [Accepted: 06/07/2002] [Indexed: 11/09/2022] Open
Abstract
Previous biochemical studies suggested that HIV-1-encoded Vpr may kill cells through an effect on the adenine nucleotide translocase (ANT), thereby causing mitochondrial membrane permeabilization (MMP). Here, we show that Vpr fails to activate caspases in conditions in which it induces cell killing. The knock-out of essential caspase-activators (Apaf-1 or caspase-9) or the knock-out of a mitochondrial caspase-independent death effector (AIF) does not abolish Vpr-mediated killing. In contrast, the cytotoxic effects of Vpr are reduced by transfection-enforced overexpression of two MMP-inhibitors, namely the endogenous protein Bcl-2 or the cytomegalovirus-encoded ANT-targeted protein vMIA. Vpr, which can elicit MMP through a direct effect on mitochondria, and HIV-1-Env, which causes MMP through an indirect pathway, exhibit additive (but not synergic) cytotoxic effects. In conclusion, it appears that Vpr induces apoptosis through a caspase-independent mitochondrial pathway.
Collapse
Affiliation(s)
- T Roumier
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Kominsky DJ, Bickel RJ, Tyler KL. Reovirus-induced apoptosis requires mitochondrial release of Smac/DIABLO and involves reduction of cellular inhibitor of apoptosis protein levels. J Virol 2002; 76:11414-24. [PMID: 12388702 PMCID: PMC136770 DOI: 10.1128/jvi.76.22.11414-11424.2002] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many viruses belonging to diverse viral families with differing structure and replication strategies induce apoptosis both in cultured cells in vitro and in tissues in vivo. Despite this fact, little is known about the specific cellular apoptotic pathways induced during viral infection. We have previously shown that reovirus-induced apoptosis of HEK cells is initiated by death receptor activation but requires augmentation by mitochondrial apoptotic pathways for its maximal expression. We now show that reovirus infection of HEK cells is associated with selective cytosolic release of the mitochondrial proapoptotic factors cytochrome c and Smac/DIABLO, but not the release of apoptosis-inducing factor. Release of these factors is not associated with loss of mitochondrial transmembrane potential and is blocked by overexpression of Bcl-2. Stable expression of caspase-9b, a dominant-negative form of caspase-9, blocks reovirus-induced caspase-9 activation but fails to significantly reduce activation of the key effector caspase, caspase-3. Smac/DIABLO enhances apoptosis through its action on cellular inhibitor of apoptosis proteins (IAPs). Reovirus infection is associated with selective down-regulation of cellular IAPs, including c-IAP1, XIAP, and survivin, effects that are blocked by Bcl-2 expression, establishing the dependence of IAP down-regulation on mitochondrial events. Taken together, these results are consistent with a model in which Smac/DIABLO-mediated inhibition of IAPs, rather than cytochrome c-mediated activation of caspase-9, is the key event responsible for mitochondrial augmentation of reovirus-induced apoptosis. These studies provide the first evidence for the association of Smac/DIABLO with virus-induced apoptosis.
Collapse
Affiliation(s)
- Douglas J Kominsky
- Department of Neurology, University of Colorado Health Science Center, Denver, Colorado 80262, USA
| | | | | |
Collapse
|
180
|
Wortman B, Darbinian N, Sawaya BE, Khalili K, Amini S. Evidence for regulation of long terminal repeat transcription by Wnt transcription factor TCF-4 in human astrocytic cells. J Virol 2002; 76:11159-65. [PMID: 12368361 PMCID: PMC136655 DOI: 10.1128/jvi.76.21.11159-11165.2002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Wnt signaling pathway plays an important role in neural cell development and function. The key components of this pathway, beta-catenin and its partner TCF-4/LEF-1, exert their effects on transcription by entering the nuclei, where they associate with the TCF-4/LEF-1 DNA motif positioned in the promoters of several important genes. Here we examined the role of TCF-4 upon transcription of the human immunodeficiency virus type 1 (HIV-1) promoter in human astrocytic cells. Our results showed that expression of TCF-4 in human astrocytic cells (U-87MG cells) decreased the basal and Tat-mediated transcription of the HIV-1 long terminal repeat (LTR). Results from promoter deletion studies revealed that the promoter sequence of the LTR with no classical binding motif for TCF-4/LEF-1, which spans positions -80 to +80 of the LTR, remained responsive to down-regulation by TCF-4. Noticeably, removal of the sequences between positions -80 and -68 decreased the negative effect of TCF-4 on viral gene transcription. A mutant variant of TCF-4 with no binding site for beta-catenin was able to down-regulate LTR transcription, suggesting that beta-catenin may not be directly involved in the observed regulatory events. Results from the glutathione S-transferase pull-down assay as well as the combined immunoprecipitation and Western blot analysis of protein extract from U-87MG cells revealed an interaction of Tat with TCF-4. Subcellular examination of TCF-4 and Tat in cells expressing either protein alone showed a predominantly nuclear accumulation of these proteins. However, in cells which coexpressed both TCF-4 and Tat, significant levels of these proteins were found in the cytoplasm. All together, these observations provide evidence for the cooperative interaction of TCF-4, the important transcription factor of the Wnt pathway, with Tat; this interaction may determine the level of viral gene transcription in human astrocytic cells.
Collapse
Affiliation(s)
- Bethany Wortman
- Center for Neurovirology and Cancer Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | | | | | | | | |
Collapse
|
181
|
Muthumani K, Hwang DS, Desai BM, Zhang D, Dayes N, Green DR, Weiner DB. HIV-1 Vpr induces apoptosis through caspase 9 in T cells and peripheral blood mononuclear cells. J Biol Chem 2002; 277:37820-31. [PMID: 12095993 DOI: 10.1074/jbc.m205313200] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human immunodeficiency virus, type 1 (HIV-1), vpr gene encodes a 14-kDa virion-associated protein, which exhibits significant effects on human cells. One important property of Vpr is its ability to induce apoptosis during infection. Apoptotic induction is likely to play a role in the pathogenesis of AIDS. However, the pathway of apoptosis is not clearly defined. In this report we investigate the mechanism of apoptosis induced by HIV-1 Vpr using a Vpr pseudotype viral infection system or adeno delivery of Vpr in primary human lymphoid cells and T-cells. With either vector, HIV-1 Vpr induced cell cycle arrest at the G(2)/M phase and apoptosis in lymphoid target cells. Furthermore, we observed that with both vectors, caspase 9, but not caspase 8, was activated following infection of human peripheral blood mononuclear cell with either Vpr-positive HIV virions or adeno-delivered Vpr. Activation of the caspase 9 pathway resulted in caspase 3 activation and apoptosis in human primary cells. These effects were coincident with the disruption of the mitochondrial transmembrane potential and induction of cytochrome c release by Vpr. The Vpr-induced signaling pathway did not induce CD95 or CD95L expression. Bcl-2 overexpressing cells succumb to Vpr-induced apoptosis. These studies illustrate that Vpr induces a mitochondria-dependent apoptotic pathway that is distinct from apoptosis driven by the Fas-FasL pathway.
Collapse
Affiliation(s)
- Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
182
|
Capano M, Crompton M. Biphasic translocation of Bax to mitochondria. Biochem J 2002; 367:169-78. [PMID: 12097139 PMCID: PMC1222873 DOI: 10.1042/bj20020805] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2002] [Revised: 07/01/2002] [Accepted: 07/04/2002] [Indexed: 11/17/2022]
Abstract
Using green fluorescent protein-tagged Bax, we demonstrate that Bax is sequestered from the cytosol of cardiomyocytes in two distinct phases following the induction of apoptosis with staurosporine. In the first phase, lasting several hours, Bax removal from the cytosol was relatively small. In the second phase, Bax was very largely removed from the cytosol and sequestered into large aggregates associated with the mitochondria. To test which of the phases involved cytochrome c release, cells were transfected with a red fluorescent protein-cytochrome c fusion. The cytochrome c fusion protein was accumulated by mitochondria of healthy cells and was released by staurosporine in phase 1. When green fluorescent protein-Bax was immunoprecipitated from extracts of cells in phase 1 and phase 2, the voltage-dependent anion channel (mitochondrial outer membrane) and the adenine nucleotide translocase (mitochondrial inner membrane) were also precipitated. These data support a two-phase model of Bax translocation in which Bax targets the mitochondrial intermembrane contact sites and releases cytochrome c in the first phase, and is then packaged into large aggregates on mitochondria in the second.
Collapse
Affiliation(s)
- Michela Capano
- Department of Biochemistry and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | | |
Collapse
|
183
|
García-Ruiz C, Colell A, Morales A, Calvo M, Enrich C, Fernández-Checa JC. Trafficking of ganglioside GD3 to mitochondria by tumor necrosis factor-alpha. J Biol Chem 2002; 277:36443-8. [PMID: 12118012 DOI: 10.1074/jbc.m206021200] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interaction of mitochondria with proapoptotic proteins activates apoptosis pathways. Previous findings have identified ganglioside GD3 (GD3) as an emerging apoptotic lipid intermediate that targets mitochondria in response to death signals. Using immunoelectron and laser scanning confocal microscopy, we characterize the trafficking of GD3 to mitochondria in response to tumor necrosis factor-alpha (TNF-alpha) in rat hepatocytes. In control hepatocytes, GD3 is present predominantly at the plasma membrane as well as in the endosomal/Golgi network, as verified by its colocalization with the asialoglycoprotein receptor. Following TNF-alpha exposure, GD3 undergoes a rapid cellular redistribution with a gradual loss from the plasma membrane before its colocalization with mitochondria. This process is mimicked by acidic sphingomyelinase and ionizing radiation but not by neutral sphingomyelinase or staurosporin. TNF-alpha stimulated the colocalization of GD3 with early and late endosomal markers, Rab 5 and Rab 7, whereas perturbation of plasma membrane cholesterol or actin cytoskeleton or inhibition of glucosylceramide synthase prevented the trafficking of GD3 to mitochondria. Finally, prevention of the TNF-alpha-stimulated neosynthesis of GD3, cyclosporin A, and latrunculin A or filipin protected sensitized hepatocytes from TNF-alpha-mediated cell death. Thus, the intracellular redistribution and mitochondrial targeting of GD3 during TNF-alpha signaling occurs through actin cytoskeleton vesicular trafficking and contributes to TNF-alpha-mediated hepatocellular cell death.
Collapse
Affiliation(s)
- Carmen García-Ruiz
- Liver Unit, Instituto de Malalties Digestives, Hospital Clinic i Provincial, Barcelona, 08036, Spain
| | | | | | | | | | | |
Collapse
|
184
|
D'Agostino DM, Ranzato L, Arrigoni G, Cavallari I, Belleudi F, Torrisi MR, Silic-Benussi M, Ferro T, Petronilli V, Marin O, Chieco-Bianchi L, Bernardi P, Ciminale V. Mitochondrial alterations induced by the p13II protein of human T-cell leukemia virus type 1. Critical role of arginine residues. J Biol Chem 2002; 277:34424-33. [PMID: 12093802 DOI: 10.1074/jbc.m203023200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human T-cell leukemia virus type 1 encodes a number of "accessory" proteins of unclear function; one of these proteins, p13(II), is targeted to mitochondria and disrupts mitochondrial morphology. The present study was undertaken to unravel the function of p13(II) through (i) determination of its submitochondrial localization and sequences required to alter mitochondrial morphology and (ii) an assessment of the biophysical and biological properties of synthetic peptides spanning residues 9-41 (p13(9-41)), which include the amphipathic mitochondrial-targeting sequence of the protein. p13(9-41) folded into an alpha helix in micellar environments. Fractionation and immunogold labeling indicated that full-length p13(II) accumulates in the inner mitochondrial membrane. p13(9-41) induced energy-dependent swelling of isolated mitochondria by increasing inner membrane permeability to small cations (Na(+), K(+)) and released Ca(2+) from Ca(2+)-preloaded mitochondria. These effects as well as the ability of full-length p13(II) to alter mitochondrial morphology in cells required the presence of four arginines, forming the charged face of the targeting signal. The mitochondrial effects of p13(9-41) were insensitive to cyclosporin A, suggesting that full-length p13(II) might alter mitochondrial permeability through a permeability transition pore-independent mechanism, thus distinguishing it from the mitochondrial proteins Vpr and X of human immunodeficiency virus type 1 and hepatitis B virus, respectively.
Collapse
Affiliation(s)
- Donna M D'Agostino
- Department of Oncology and Surgical Sciences, University of Padova, via Gattamelata 64, 35128 Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Machida K, Hayashi Y, Osada H. A novel adenine nucleotide translocase inhibitor, MT-21, induces cytochrome c release by a mitochondrial permeability transition-independent mechanism. J Biol Chem 2002; 277:31243-8. [PMID: 12063261 DOI: 10.1074/jbc.m204564200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The release of cytochrome c from mitochondria is a critical step during apoptosis. In order to study this process, we have used a synthetic compound, MT-21, that is able to initiate release of cytochrome c from isolated mitochondria. We demonstrate that MT-21 significantly inhibits ADP transport activity in mitochondria and reduces binding of the adenine nucleotide translocase (ANT) to a phenylarsine oxide affinity matrix. These results suggest that ANT, one of the components of the mitochondrial permeability transition (PT) pore, is the molecular target for MT-21. In agreement with this, the MT-21-induced cytochrome c release was effectively inhibited in the presence of ANT ligands, and MT-21 could dissociate ANT from a complex with a glutathione S-transferase-cyclophilin D fusion protein. Interestingly, we also found that specific inhibitors of ANT such as MT-21 and atractyloside could induce cytochrome c release without mitochondrial swelling and that this event was highly dependent on the presence of Mg(2+). These results suggest that although ANT resides in the mitochondrial inner membrane, specific ANT inhibitors can induce cytochrome c release without having an effect on inner membrane permeability. Therefore, MT-21 can be a powerful tool for studying the mechanism of PT-independent cytochrome c release from mitochondria.
Collapse
Affiliation(s)
- Kiyotaka Machida
- Antibiotics Laboratory, RIKEN, Hirosawa 2-1, Saitama 351-0198, Japan
| | | | | |
Collapse
|
186
|
Castedo M, Roumier T, Blanco J, Ferri KF, Barretina J, Tintignac LA, Andreau K, Perfettini JL, Amendola A, Nardacci R, Leduc P, Ingber DE, Druillennec S, Roques B, Leibovitch SA, Vilella-Bach M, Chen J, Este JA, Modjtahedi N, Piacentini M, Kroemer G. Sequential involvement of Cdk1, mTOR and p53 in apoptosis induced by the HIV-1 envelope. EMBO J 2002; 21:4070-80. [PMID: 12145207 PMCID: PMC126138 DOI: 10.1093/emboj/cdf391] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Syncytia arising from the fusion of cells expressing the HIV-1-encoded Env gene with cells expressing the CD4/CXCR4 complex undergo apoptosis following the nuclear translocation of mammalian target of rapamycin (mTOR), mTOR-mediated phosphorylation of p53 on Ser15 (p53(S15)), p53-dependent upregulation of Bax and activation of the mitochondrial death pathway. p53(S15) phosphorylation is only detected in syncytia in which nuclear fusion (karyogamy) has occurred. Karyogamy is secondary to a transient upregulation of cyclin B and a mitotic prophase-like dismantling of the nuclear envelope. Inhibition of cyclin-dependent kinase-1 (Cdk1) prevents karyogamy, mTOR activation, p53(S15) phosphorylation and apoptosis. Neutralization of p53 fails to prevent karyogamy, yet suppresses apoptosis. Peripheral blood mononuclear cells from HIV-1-infected patients exhibit an increase in cyclin B and mTOR expression, correlating with p53(S15) phosphorylation and viral load. Cdk1 inhibition prevents the death of syncytia elicited by HIV-1 infection of primary CD4 lymphoblasts. Thus, HIV-1 elicits a pro-apoptotic signal transduction pathway relying on the sequential action of cyclin B-Cdk1, mTOR and p53.
Collapse
Affiliation(s)
| | | | - Julià Blanco
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | | | - Jordi Barretina
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | | | | | | | - Alessandra Amendola
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Roberta Nardacci
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Philip Leduc
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Donald E. Ingber
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Sabine Druillennec
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Bernard Roques
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | | | - Montserrat Vilella-Bach
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Jie Chen
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - José A. Este
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | | | - Mauro Piacentini
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| | - Guido Kroemer
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif,
Unité de Pharmacochimie Moléculaire et Structurale, INSERM U266–CNRS UMR860, Université René Descartes (Paris V), F-75005 Paris, France, Institut de Recerca de la SIDA-Caixa, Laboratori de Retrovirologia, Hospital Universitari Germans Trias i Pujol, Universitat Autónoma de Barcelona, Ctra Canyet s/n, 08916 Badalona, Catalonia, Spain, Istituto Nazionale Malattie Infettive ‘L. Spallanzani’, Rome 00149, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy, Departments of Surgery and Pathology, Children’s Hospital and Harvard Medical School, Enders 1007, 300 Longwood Avenue, Boston, MA 02115 and Department of Cell and Structural Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA Corresponding author e-mail:
| |
Collapse
|
187
|
Pilon AA, Lum JJ, Sanchez-Dardon J, Phenix BN, Douglas R, Badley AD. Induction of apoptosis by a nonnucleoside human immunodeficiency virus type 1 reverse transcriptase inhibitor. Antimicrob Agents Chemother 2002; 46:2687-91. [PMID: 12121958 PMCID: PMC127389 DOI: 10.1128/aac.46.8.2687-2691.2002] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inhibition of human immunodeficiency virus type 1 reverse transcriptase (RT) by both nucleoside and nonnucleoside RT inhibitors profoundly inhibits virus replication. Nucleoside RT inhibitors are known to be toxic, but there is little information regarding the toxicities of nonnucleoside RT inhibitors (NNRTI). We demonstrate that efavirenz (an NNRTI) induces caspase- and mitochondrion-dependent apoptosis of Jurkat T cells and human peripheral blood mononuclear cells. The clinical relevance of these observations is not yet clear.
Collapse
Affiliation(s)
- A A Pilon
- HIV Research Laboratory, Ottawa Health Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | | | | | | | | | | |
Collapse
|
188
|
Lefèbvre L, Ciminale V, Vanderplasschen A, D'Agostino D, Burny A, Willems L, Kettmann R. Subcellular localization of the bovine leukemia virus R3 and G4 accessory proteins. J Virol 2002; 76:7843-54. [PMID: 12097596 PMCID: PMC136348 DOI: 10.1128/jvi.76.15.7843-7854.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2002] [Accepted: 04/23/2002] [Indexed: 11/20/2022] Open
Abstract
Bovine leukemia virus (BLV) is a complex retrovirus that belongs to the Deltaretrovirus genus, which also includes Human T-cell leukemia virus type 1 (HTLV-1). Both viruses contain an X region coding for at least four proteins: Tax and Rex, which are involved in transcriptional and posttranscriptional regulation, respectively, and the accessory proteins R3 and G4 (for BLV) and p12(I), p13(II), and p30(II) (for HTLV-1). The present study was aimed at characterizing the subcellular localization of BLV R3 and G4. The results of immunofluorescence experiments on transfected HeLa Tat cells demonstrated that R3 is located in the nucleus and in cellular membranes, as previously reported for HTLV-1 p12(I). In contrast, G4, like p13(II), is localized both in the nucleus and in mitochondria. In addition, we have shown that G4 harbors a mitochondrial targeting signal consisting of a hydrophobic region and an amphipathic alpha-helix. Thus, despite a lack of significant primary sequence homology, R3 and p12(I) and G4 and p13(II) exhibit similar targeting properties, suggesting possible overlap in their functional properties.
Collapse
Affiliation(s)
- Laurent Lefèbvre
- Faculty of Agronomy, Gembloux, University of Liège, Liège, Belgium
| | | | | | | | | | | | | |
Collapse
|
189
|
Wecker K, Morellet N, Bouaziz S, Roques BP. NMR structure of the HIV-1 regulatory protein Vpr in H2O/trifluoroethanol. Comparison with the Vpr N-terminal (1-51) and C-terminal (52-96) domains. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:3779-88. [PMID: 12153575 DOI: 10.1046/j.1432-1033.2002.03067.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human immunodeficiency virus type 1, HIV-1, genome encodes a highly conserved regulatory gene product, Vpr (96 amino acids), which is incorporated into virions in quantities equivalent to those of the viral Gag protein. In infected cells, Vpr is believed to function during the early stages of HIV-1 replication (such as transcription of the proviral genome and migration of preintegration nuclear complex), blocks cells in G2 phase and triggers apoptosis. Vpr also plays a critical role in long-term AIDS disease by inducing viral infection in nondividing cells such as monocytes and macrophages. To gain deeper insight of the structure-function relationship of Vpr, the intact protein (residues 1-96) was synthesized. Its three-dimensional structure was analysed using circular dichroism and two-dimensional 1H- and 15N-NMR and refined by restrained molecular dynamics. In addition, 15N relaxation parameters (T1, T2) and heteronuclear 1H-15N NOEs were measured. The structure of the protein is characterized by a well-defined gamma turn(14-16)-alpha helix(17-33)-turn(34-36), followed by a alpha helix(40-48)-loop(49-54)-alpha helix(55-83) domain and ends with a very flexible C-terminal sequence. This structural determination of the whole intact Vpr molecule provide insights into the biological role played by this protein during the virus life cycle, as such amphipathic helices are believed to be involved in protein-lipid bilayers, protein-protein and/or protein-nucleic acid interactions.
Collapse
Affiliation(s)
- K Wecker
- Département de Pharmacochimie Moléculaire et Structurale, INSERM U266 CNRS UMR 8600, UFR des Sciences Pharmaceutiques et Biologiques, Paris, France.
| | | | | | | |
Collapse
|
190
|
Muthumani K, Zhang D, Hwang DS, Kudchodkar S, Dayes NS, Desai BM, Malik AS, Yang JS, Chattergoon MA, Maguire HC, Weiner DB. Adenovirus encoding HIV-1 Vpr activates caspase 9 and induces apoptotic cell death in both p53 positive and negative human tumor cell lines. Oncogene 2002; 21:4613-25. [PMID: 12096338 DOI: 10.1038/sj.onc.1205549] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2002] [Revised: 03/20/2002] [Accepted: 03/27/2002] [Indexed: 11/08/2022]
Abstract
The targeted delivery of genes whose products arrest the cell cycle and/or induce apoptosis represent an important tool for the understanding and controlling forms of unregulated cell growth. The vpr gene product of HIV-1 has been reported to interfere with cell growth and induce apoptosis, but the mechanism of its action is not clearly understood. In order to study these important properties of Vpr, we created a recombinant adenovirus H5.010CMV-vpr (adCMV-vpr) as a tool to deliver the vpr gene to various cell lines to examine its biology. Vpr protein expression was confirmed by Western blot analysis in adCMV-vpr infected cells. We tested the effects of adCMV-vpr on cell growth of several tumor cell lines. Infection of both p53 positive and p53 deficient tumor cell lines with adCMV-vpr resulted in dramatic induction of cell death in short-term assays. We observed that apoptosis was induced through the mitochondrial pathway as we observed changes in the cytochrome c content accompanied by caspase 9 activation. As Bcl-2 is reported to interfere with apoptosis through the mitochondrial pathway, we examined the effect of adCMV-vpr in Bcl-2 over expressing cell lines. We observed that Bcl-2 overexpression does not inhibit adCMV-vpr induced apoptosis. The properties of adCMV-vpr inducing apoptosis through caspase 9 in a p53 pathway independent manner suggest that this is an important reagent. Such a vector may give insight into approaches designed to limit the growth of pathogenic human cells.
Collapse
Affiliation(s)
- Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Müller A, Rassow J, Grimm J, Machuy N, Meyer TF, Rudel T. VDAC and the bacterial porin PorB of Neisseria gonorrhoeae share mitochondrial import pathways. EMBO J 2002; 21:1916-29. [PMID: 11953311 PMCID: PMC125974 DOI: 10.1093/emboj/21.8.1916] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The human pathogen Neisseria gonorrhoeae induces host cell apoptosis during infection by delivering the outer membrane protein PorB to the host cell's mitochondria. PorB is a pore-forming beta-barrel protein sharing several features with the mitochondrial voltage-dependent anion channel (VDAC), which is involved in the regulation of apoptosis. Here we show that PorB of pathogenic Neisseria species produced by host cells is efficiently targeted to mitochondria. Imported PorB resides in the mitochondrial outer membrane and forms multimers with similar sizes as in the outer bacterial membrane. The mitochondria completely lose their membrane potential, a characteristic previously observed in cells infected with gonococci or treated with purified PorB. Closely related bacterial porins of non-pathogenic Neisseria mucosa or Escherichia coli remain in the cytosol. Import of PorB into mitochondria in vivo is independent of a linear signal sequence. Insertion of PorB into the mitochondrial outer membrane in vitro depends on the activity of Tom5, Tom20 and Tom40, but is independent of Tom70. Our data show that human VDAC and bacterial PorB are imported into mitochondria by a similar mechanism.
Collapse
Affiliation(s)
| | - Joachim Rassow
- Max Planck Institute for Infection Biology, Department of Molecular Biology, Schumannstrasse 21/22, D-10117 Berlin,
University of Hohenheim, Department of Microbiology, Garbenstrasse 30, D-70593 Stuttgart-Hohenheim and Max Delbrück Centrum für Molekulare Medizin, Abteilung Zellbiologie, Robert-Rössle-Strasse 10, D-13092 Berlin, Germany Corresponding author e-mail:
| | - Jan Grimm
- Max Planck Institute for Infection Biology, Department of Molecular Biology, Schumannstrasse 21/22, D-10117 Berlin,
University of Hohenheim, Department of Microbiology, Garbenstrasse 30, D-70593 Stuttgart-Hohenheim and Max Delbrück Centrum für Molekulare Medizin, Abteilung Zellbiologie, Robert-Rössle-Strasse 10, D-13092 Berlin, Germany Corresponding author e-mail:
| | | | - Thomas F. Meyer
- Max Planck Institute for Infection Biology, Department of Molecular Biology, Schumannstrasse 21/22, D-10117 Berlin,
University of Hohenheim, Department of Microbiology, Garbenstrasse 30, D-70593 Stuttgart-Hohenheim and Max Delbrück Centrum für Molekulare Medizin, Abteilung Zellbiologie, Robert-Rössle-Strasse 10, D-13092 Berlin, Germany Corresponding author e-mail:
| | | |
Collapse
|
192
|
Patel CA, Mukhtar M, Harley S, Kulkosky J, Pomerantz RJ. Lentiviral expression of HIV-1 Vpr induces apoptosis in human neurons. J Neurovirol 2002; 8:86-99. [PMID: 11935461 DOI: 10.1080/13550280290049552] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Our recent studies have demonstrated that extracellular, recombinant human immunodeficiency virus type I (HIV-1) Vpr protein is highly neurotoxic in the microenvironment of differentiated mature human neurons and undifferentiated neuronal precursors. Although most of the direct neurotoxic effects of HIV-1 have been attributed previously to the envelope gene product, gp120, and the Tat regulatory protein, it was demonstrated that Vpr protein caused apoptosis comparable to that induced by gp120 protein in a dose-dependent manner in the neuronal system. Having observed the neurocytopathic effects of extracellular Vpr protein previously, the effects of virally expressed Vpr on nondividing, terminally differentiated human neurons were investigated. An HIV-1-based three-plasmid expression vector system was utilized to study the effects of intracellularly expressed Vpr. These virion preparations were then used to transduce neurons generated from the human neuronal precursor NT2 cell-line. Intracellularly expressed Vpr induced apoptosis within terminally differentiated neurons, as demonstrated by TUNEL assays. Additionally, virions lacking Vpr expression did not significantly induce apoptosis within these neurons. These results suggest that HIV-1 Vpr may also be leading directly to selective neurotoxicity through intracellular expression. Furthermore, human apoptosis gene microarray comparisons exhibited an up-regulation of Bcl-2-related mRNA, as well as other apoptosis genes involved in the mitochondrial apoptotic pathway, for the Vpr-transduced neuronal cells, when compared to Vpr-negative controls. Thus, Vpr delivered intracellularly, as well as extracellularly, is involved in the induction of significant neuronal apoptosis and may be one of the molecular mechanisms in HIV-1-induced encephalopathy.
Collapse
Affiliation(s)
- Charvi A Patel
- The Dorrance H. Hamilton Laboratories, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|
193
|
Vieira HLA, Boya P, Cohen I, El Hamel C, Haouzi D, Druillenec S, Belzacq AS, Brenner C, Roques B, Kroemer G. Cell permeable BH3-peptides overcome the cytoprotective effect of Bcl-2 and Bcl-X(L). Oncogene 2002; 21:1963-77. [PMID: 11960369 DOI: 10.1038/sj.onc.1205270] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2001] [Revised: 12/13/2001] [Accepted: 12/19/2001] [Indexed: 11/08/2022]
Abstract
Peptides corresponding to the BH3 domains of Bax (BaxBH3) or Bcl-2 (Bcl2BH3) are potent inducers of apoptosis when fused to the Atennapedia plasma membrane translocation domain (Ant). BaxBH3Ant and Bcl2BH3Ant caused a mitochondrial membrane permeabilization (MMP) and apoptosis, via a mechanism that was not inhibited by overexpressed Bcl-2 or Bcl-X(L), yet partially inhibited by cyclosporin A (CsA), an inhibitor of the mitochondrial permeability transition pore. When added to isolated mitochondria, BaxBH3 and Bcl2BH3 induced MMP, which was inhibited by CsA. However, Bcl-2 or Bcl-X(L) failed to inhibit MMP induced by BaxBH3 and Bc2BH3 in vitro, while they efficiently suppressed the induction of MMP by the Vpr protein (from human immunodeficiency virus-1), a ligand of the adenine nucleotide translocator (ANT). BaxBH3 but not Bcl2BH3 was found to interact with ANT, and only BaxBH3 (not Bcl2BH3) permeabilized ANT proteoliposomes and induced ANT to form non-specific channels in electrophysiological experiments. In contrast, both BaxBH3 and Bcl2BH3 were able to stimulate channel formation by recombinant Bax protein. Thus, BaxBH3 might induce MMP via an action on at least two targets, ANT and Bax-like proteins. In contrast, Bcl2BH3 would elicit MMP in an ANT-independent fashion. In purified mitochondria, two ligands of ANT, bongkrekic acid and the protein vMIA from cytomegalovirus, failed to prevent MMP induced by BaxBH3 or Bcl2BH3. In conclusion, BaxBH3 and Bcl2BH3 induce MMP and apoptosis through a mechanism which overcomes cytoprotection by Bcl-2 and Bcl-X(L).
Collapse
Affiliation(s)
- Helena L A Vieira
- Centre National de la Recherche Scientifique, UMR1599, Institut Gustave Roussy, 39 rue Camille-Desmoulins, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Abstract
Mitochondria play a critical role in initiating both apoptotic and necrotic cell death. A major player in this process is the mitochondrial permeability transition pore (MPTP), a non-specific pore, permeant to any molecule of < 1.5 kDa, that opens in the inner mitochondrial membrane under conditions of elevated matrix [Ca(2+)], especially when this is accompanied by oxidative stress and depleted adenine nucleotides. Opening of the MPTP causes massive swelling of mitochondria, rupture of the outer membrane and release of intermembrane components that induce apoptosis. In addition mitochondria become depolarised causing inhibition of oxidative phosphorylation and stimulation of ATP hydrolysis. Pore opening is inhibited by cyclosporin A analogues with the same affinity as they inhibit the peptidyl-prolyl cis-trans isomerase activity of mitochondrial cyclophilin (CyP-D). These data and the observation that different ligands of the adenine nucleotide translocase (ANT) can either stimulate or inhibit pore opening led to the proposal that the MPTP is formed by a Ca-triggered conformational change of the ANT that is facilitated by the binding of CyP-D. Our model is able to explain the mode of action of a wide range of known modulators of the MPTP that exert their effects by changing the binding affinity of the ANT for CyP-D, Ca(2+) or adenine nucleotides. The extensive evidence for this model from our own and other laboratories is presented, including reconstitution studies that demonstrate the minimum configuration of the MPTP to require neither the voltage activated anion channel (VDAC or porin) nor any other outer membrane protein. However, other proteins including Bcl-2, BAX and virus-derived proteins may interact with the ANT to regulate the MPTP. Recent data suggest that oxidative cross-linking of two matrix facing cysteine residues on the ANT (Cys(56) and Cys(159)) plays a key role in regulating the MPTP. Adenine nucleotide binding to the ANT is inhibited by Cys(159) modification whilst oxidation of Cys(56) increases CyP-D binding to the ANT, probably at Pro(61).
Collapse
|
195
|
Jan G, Belzacq AS, Haouzi D, Rouault A, Métivier D, Kroemer G, Brenner C. Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria. Cell Death Differ 2002; 9:179-88. [PMID: 11840168 DOI: 10.1038/sj.cdd.4400935] [Citation(s) in RCA: 249] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2001] [Revised: 07/26/2001] [Accepted: 08/03/2001] [Indexed: 12/13/2022] Open
Abstract
The genus Propionibacterium is composed of dairy and cutaneous bacteria which produce short-chain fatty acids (SCFA), mainly propionate and acetate, by fermentation. Here, we show that P. acidipropionici and freudenreichii, two species which can survive in the human intestine, can kill two human colorectal carcinoma cell lines by apoptosis. Propionate and acetate were identified as the major cytotoxic components secreted by the bacteria. Bacterial culture supernatants as well as pure SCFA induced typical signs of apoptosis including a loss of mitochondrial transmembrane potential, the generation of reactive oxygen species, caspase-3 processing, and nuclear chromatin condensation. The oncoprotein Bcl-2, which is known to prevent apoptosis via mitochondrial effects, and the cytomegalovirus-encoded protein vMIA, which inhibits apoptosis and interacts with the mitochondrial adenine nucleotide translocator (ANT), both inhibited cell death induced by propionibacterial SCFA, suggesting that mitochondria and ANT are involved in the cell death pathway. Accordingly, propionate and acetate induced mitochondrial swelling when added to purified mitochondria in vitro. Moreover, they specifically permeabi-lize proteoliposomes containing ANT, indicating that ANT can be a critical target in SCFA-induced apoptosis. We suggest that propionibacteria could constitute probiotics efficient in digestive cancer prophylaxis via their ability to produce apoptosis-inducing SCFA.
Collapse
Affiliation(s)
- G Jan
- Institut National de la Recherche Agronomique, UR 121, Laboratoire de Recherches de Technologie Laitière, 35042 Rennes Cedex, France
| | | | | | | | | | | | | |
Collapse
|
196
|
Abstract
Alteration of mitochondrial membrane permeability is a central mechanism leading invariably to cell death, which results, at least in part, from the opening of the permeability transition pore complex (PTPC). Indeed, extended PTPC opening is sufficient to trigger an increase in mitochondrial membrane permeability and apoptosis. Among the various PTPC components, the adenine nucleotide translocator (ANT) appears to act as a bi-functional protein which, on the one hand, contributes to a crucial step of aerobic energy metabolism, the ADP/ATP translocation, and on the other hand, can be converted into a pro-apoptotic pore under the control of onco- and anti-oncoproteins from the Bax/Bcl-2 family. In this review, we will discuss recent advances in the cooperation between ANT and Bax/Bcl-2 family members, the multiplicity of agents affecting ANT pore function and the putative role of ANT isoforms in apoptosis control.
Collapse
Affiliation(s)
- Anne-Sophie Belzacq
- Centre national de la recherche scientifique, UMR 6022, université de technologie de Compiègne, Royallieu, BP20529, 60205 Compiègne, France
| | | | | | | |
Collapse
|
197
|
Crompton M, Barksby E, Johnson N, Capano M. Mitochondrial intermembrane junctional complexes and their involvement in cell death. Biochimie 2002; 84:143-52. [PMID: 12022945 DOI: 10.1016/s0300-9084(02)01368-8] [Citation(s) in RCA: 198] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mitochondria establish contact sites between the inner and outer membranes. The contact sites are held together by junctional complexes of the adenine nucleotide translocase (ANT; inner membrane) and the voltage-dependent anion channel (VDAC; outer membrane). The junctional complexes act as multifunctional recruitment centres, binding a range of proteins according to the function to be executed. Some of these, involving kinases and enzymes of lipid transfer, are readily understood as ongoing functions in energy and lipid metabolism. But the roles of other proteins recruited to the junctional complexes are less well defined. Here, we focus on the complexes formed with Bax and with cyclophilin-D, and their possible roles in apoptotic and necrotic cell death. We have isolated both types of complexes using glutathione-S-transferase fusion proteins of Bax and of cyclophilin-D. The VDAC/ANT/cyclophilin-D complex reconstitutes Ca(2+)- and cyclosporin A-sensitive permeability transition pore activity when incorporated into proteoliposomes. The complex forms readily in the absence of factors required for pore opening in isolated mitochondria, suggesting that these factors act on the preexisting complex, rather than drive its assembly, and that the complex is a physiological entity in healthy cells.
Collapse
Affiliation(s)
- Martin Crompton
- Department of Biochemistry and Molecular Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | | | | | | |
Collapse
|
198
|
Phenix BN, Badley AD. Influence of mitochondrial control of apoptosis on the pathogenesis, complications and treatment of HIV infection. Biochimie 2002; 84:251-64. [PMID: 12022956 DOI: 10.1016/s0300-9084(02)01378-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
HIV infection is inexorably linked with disordered regulation of apoptosis, and consequent alterations in mitochondrial homeostasis, resulting in CD4 T cell death and enhanced susceptibility to opportunistic infections and malignancies. Effective treatment of HIV reverses the changes in mitochondrial homeostasis and apoptosis, and enhances immunocompetence. This review will summarize current knowledge of: i) the associations of apoptosis with HIV disease progression; ii) mechanisms of enhanced apoptosis in HIV infection; iii) putative role of apoptosis in HIV complications; iv) direct effects of HIV therapies on mitochondria and apoptosis; and finally v) treatment strategies for HIV based upon modifying the apoptotic response.
Collapse
Affiliation(s)
- Barbara N Phenix
- Ottawa Health Research Institute, University of Ottawa, 501 Smyth Road, Ottawa. Ontario, Canada K1H 8L6
| | | |
Collapse
|
199
|
Wasilenko ST, Meyers AF, Vander Helm K, Barry M. Vaccinia virus infection disarms the mitochondrion-mediated pathway of the apoptotic cascade by modulating the permeability transition pore. J Virol 2001; 75:11437-48. [PMID: 11689625 PMCID: PMC114730 DOI: 10.1128/jvi.75.23.11437-11448.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Many viruses have evolved strategies that target crucial components within the apoptotic cascade. One of the best studied is the caspase 8 inhibitor, crmA/Spi-2, encoded by members of the poxvirus family. Since many proapoptotic stimuli induce apoptosis through a mitochondrion-dependent, caspase 8-independent pathway, we hypothesized that vaccinia virus would encode a mechanism to directly modulate the mitochondrial apoptotic pathway. In support of this, we observed that Jurkat cells, which undergo Fas-mediated apoptosis exclusively through the mitochondrial route, were resistant to Fas-induced death following infection with a crmA/Spi-2-deficient strain of vaccinia virus. In addition, vaccinia virus-infected cells subjected to the proapoptotic stimulus staurosporine exhibited decreased levels of both cytochrome c released from the mitochondria and caspase 3 activation. In all cases we found that the loss of the mitochondrial membrane potential, which occurs as a result of opening the multimeric permeability transition pore complex, was prevented in vaccinia virus-infected cells. Moreover, vaccinia virus infection specifically inhibited opening of the permeability transition pore following treatment with the permeability transition pore ligand atractyloside and t-butylhydroperoxide. These studies indicate that vaccinia virus infection directly impacts the mitochondrial apoptotic cascade by influencing the permeability transition pore.
Collapse
Affiliation(s)
- S T Wasilenko
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | | | | | | |
Collapse
|
200
|
Belzacq AS, El Hamel C, Vieira HL, Cohen I, Haouzi D, Métivier D, Marchetti P, Brenner C, Kroemer G. Adenine nucleotide translocator mediates the mitochondrial membrane permeabilization induced by lonidamine, arsenite and CD437. Oncogene 2001; 20:7579-87. [PMID: 11753636 DOI: 10.1038/sj.onc.1204953] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2001] [Revised: 08/03/2001] [Accepted: 09/04/2001] [Indexed: 01/31/2023]
Abstract
An increasing number of experimental chemotherapeutic agents induce apoptosis by directly triggering mitochondrial membrane permeabilization (MMP). Here we examined MMP induced by lonidamine, arsenite, and the retinoid derivative CD437. Cells overexpressing the cytomegalovirus-encoded protein vMIA, a protein which interacts with the adenine nucleotide translocator, were strongly protected against the MMP-inducing and apoptogenic effects of lonidamine, arsenite, and CD437. In a cell-free system, lonidamine, arsenite, and CD437 induced the permeabilization of ANT proteoliposomes, yet had no effect on protein-free liposomes. The ANT-dependent membrane permeabilization was inhibited by the two ANT ligands ATP and ADP, as well as by recombinant Bcl-2 protein. Lonidamine, arsenite, and CD437, added to synthetic planar lipid bilayers containing ANT, elicited ANT channel activities with clearly distinct conductance levels of 20+/-7, 100+/-30, and 47+/-7 pS, respectively. Altering the ATP/ADP gradient built up on the inner mitochondrial membrane by inhibition of glycolysis and/or oxidative phosphorylation differentially modulated the cytocidal potential of lonidamine, arsenite, and CD437. Inhibition of F(0)F(1)ATPase without glycolysis inhibition sensitized to lonidamine-induced cell death. In contrast, only the combined inhibition of glycolysis plus F(0)F(1)ATPase sensitized to arsenite-induced cell death. No sensitization to cell death induction by CD437 was achieved by glucose depletion and/or oligomycin addition. These results indicate that ANT is a target of lonidamine, arsenite, and CD437 and unravel an unexpected heterogeneity in the mode of action of these three compounds.
Collapse
Affiliation(s)
- A S Belzacq
- CNRS-UMR6022, Université de Technologie de Compiègne, BP 20529, F-60205 Compiègne, France
| | | | | | | | | | | | | | | | | |
Collapse
|