251
|
Abstract
Cytotoxic approaches to killing tumor cells, such as chemotherapeutic agents, gamma-irradiation, suicide genes or immunotherapy, have been shown to induce cell death through apoptosis. The intrinsic apoptotic pathway is activated following treatment with cytotoxic drugs, and these reactions ultimately lead to the activation of caspases, which promote cell death in tumor cells. In addition, activation of the extrinsic apoptotic pathway with death-inducing ligands leads to an increased sensitivity of tumor cells toward cytotoxic stimuli, illustrating the interplay between the two cell death pathways. In contrast, tumor resistance to cytotoxic stimuli may be due to defects in apoptotic signaling. As a result of their importance in killing cancer cells, a number of apoptotic molecules are implicated in cancer therapy. The knowledge gleaned from basic research into apoptotic pathways from cell biological, structural, biochemical, and biophysical approaches can be used in strategies to develop novel compounds that eradicate tumor cells. In addition to current drug targets, research into molecules that activate procaspase-3 directly may show the direct activation of the executioner caspase to be a powerful therapeutic strategy in the treatment of many cancers.
Collapse
Affiliation(s)
- Sarah H. MacKenzie
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - A. Clay Clark
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
252
|
Nachmias B, Mizrahi S, Elmalech M, Lazar I, Ashhab Y, Gazit R, Markel G, Ben-Yehuda D, Mandelboim O. Manipulation of NK cytotoxicity by the IAP family member Livin. Eur J Immunol 2008; 37:3467-76. [PMID: 18034418 DOI: 10.1002/eji.200636600] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Natural killer (NK) cells are part of the innate immune system, capable of killing tumor and virally infected cells. NK cells induce apoptosis in the target cell by either granule- or receptor-mediated pathways. A set of inhibitory and activation ligands governs NK cell activation. As transformed cells often attempt to evade NK cell killing, up-regulation of a potential anti-apoptotic factor should provide a survival advantage. The inhibitor of apoptosis protein (IAP) family can inhibit apoptosis induced by a variety of stimuli. We have previously described a new IAP family member, termed Livin, which has two splice variants (alpha and beta) with differential anti-apoptotic activities. In this study, we explore the ability of Livin to inhibit NK cell-induced killing. We demonstrate that Livin beta moderately protects against NK cell killing whereas Livin alpha augments killing. We show that Livin beta inhibition in Jurkat cells is apparent upon concomitant activation of an inhibitory signal, suggesting that Livin augments an extrinsic inhibitory signal rather than functioning as an independent inhibitory mechanism. Finally, we demonstrate that detection of both Livin isoforms in melanoma cells correlates with a low killing rate. To date, this is the first evidence that directly demonstrates the ability of IAP to protect against NK cell-induced apoptosis.
Collapse
Affiliation(s)
- Boaz Nachmias
- Department of Hematology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
253
|
CHENG Y, MAEDA A, GOTO Y, MATSUDA F, MIYANO T, INOUE N, SAKAMAKI K, MANABE N. Changes in Expression and Localization of X-linked Inhibitor of Apoptosis Protein (XIAP) in Follicular Granulosa Cells During Atresia in Porcine Ovaries. J Reprod Dev 2008; 54:454-9. [DOI: 10.1262/jrd.20088] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yuan CHENG
- Animal Resource Science Center, The University of Tokyo
| | - Akihisa MAEDA
- Animal Resource Science Center, The University of Tokyo
| | - Yasufumi GOTO
- Animal Resource Science Center, The University of Tokyo
| | - Fuko MATSUDA
- Animal Resource Science Center, The University of Tokyo
| | - Takashi MIYANO
- Animal Reproduction and Biotechnology, Graduate School of Agricultural Science, Kobe University
| | - Naoko INOUE
- Laboratory of Animal Morphology and Function, Graduate School of Bioagricultural Sciences, Nagoya University
| | - Kazuhiro SAKAMAKI
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University
| | - Noboru MANABE
- Animal Resource Science Center, The University of Tokyo
- Research Center for Food Safety, The University of Tokyo
| |
Collapse
|
254
|
Ribeiro PS, Kuranaga E, Tenev T, Leulier F, Miura M, Meier P. DIAP2 functions as a mechanism-based regulator of drICE that contributes to the caspase activity threshold in living cells. J Cell Biol 2007; 179:1467-80. [PMID: 18166655 PMCID: PMC2373516 DOI: 10.1083/jcb.200706027] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Accepted: 11/22/2007] [Indexed: 01/08/2023] Open
Abstract
In addition to their well-known function in apoptosis, caspases are also important in several nonapoptotic processes. How caspase activity is restrained and shut down under such nonapoptotic conditions remains unknown. Here, we show that Drosophila melanogaster inhibitor of apoptosis protein 2 (DIAP2) controls the level of caspase activity in living cells. Animals that lack DIAP2 have higher levels of drICE activity. Although diap2-deficient cells remain viable, they are sensitized to apoptosis following treatment with sublethal doses of x-ray irradiation. We find that DIAP2 regulates the effector caspase drICE through a mechanism that resembles the one of the caspase inhibitor p35. As for p35, cleavage of DIAP2 is required for caspase inhibition. Our data suggest that DIAP2 forms a covalent adduct with the catalytic machinery of drICE. In addition, DIAP2 also requires a functional RING finger domain to block cell death and target drICE for ubiquitylation. Because DIAP2 efficiently interacts with drICE, our data suggest that DIAP2 controls drICE in its apoptotic and nonapoptotic roles.
Collapse
Affiliation(s)
- Paulo S Ribeiro
- Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, London SW3 6JB, England, UK
| | | | | | | | | | | |
Collapse
|
255
|
Sun H, Nikolovska-Coleska Z, Lu J, Meagher JL, Yang CY, Qiu S, Tomita Y, Ueda Y, Jiang S, Krajewski K, Roller PP, Stuckey JA, Wang S. Design, synthesis, and characterization of a potent, nonpeptide, cell-permeable, bivalent Smac mimetic that concurrently targets both the BIR2 and BIR3 domains in XIAP. J Am Chem Soc 2007; 129:15279-94. [PMID: 17999504 PMCID: PMC2553712 DOI: 10.1021/ja074725f] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
XIAP is a central apoptosis regulator that inhibits apoptosis by binding to and inhibiting the effectors caspase-3/-7 and an initiator caspase-9 through its BIR2 and BIR3 domains, respectively. Smac protein in its dimeric form effectively antagonizes XIAP by concurrently targeting both its BIR2 and BIR3 domains. We report the design, synthesis, and characterization of a nonpeptide, cell-permeable, bivalent small-molecule (SM-164) which mimics Smac protein for targeting XIAP. Our study shows that SM-164 binds to XIAP containing both BIR domains with an IC50 value of 1.39 nM, being 300 and 7000 times more potent than its monovalent counterparts and the natural Smac AVPI peptide, respectively. SM-164 concurrently interacts with both BIR domains in XIAP and functions as an ultrapotent antagonist of XIAP in both cell-free functional and cell-based assays. SM-164 targets cellular XIAP and effectively induces apoptosis at concentrations as low as 1 nM in the HL-60 leukemia cell line. The potency of bivalent SM-164 in binding, functional, and cellular assays is 2-3 orders of magnitude higher than its corresponding monovalent Smac mimetics.
Collapse
Affiliation(s)
- Haiying Sun
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Zaneta Nikolovska-Coleska
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Jianfeng Lu
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Jennifer L. Meagher
- Life Sciences Institute, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Chao-Yie Yang
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Su Qiu
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - York Tomita
- Lombardi Cancer Center, Georgetown University Medical Center, Washington DC 20007, USA
| | - Yumi Ueda
- Lombardi Cancer Center, Georgetown University Medical Center, Washington DC 20007, USA
| | - Sheng Jiang
- Laboratory of Medicinal Chemistry, National Cancer Institute-Frederick, NIH, Frederick, Maryland 21702, USA
| | - Krzysztof Krajewski
- Laboratory of Medicinal Chemistry, National Cancer Institute-Frederick, NIH, Frederick, Maryland 21702, USA
| | - Peter P. Roller
- Laboratory of Medicinal Chemistry, National Cancer Institute-Frederick, NIH, Frederick, Maryland 21702, USA
| | - Jeanne A. Stuckey
- Life Sciences Institute, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Biological Chemistry, Biophysics Research Division, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Departments of Internal Medicine, Pharmacology and Medicinal Chemistry and Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| |
Collapse
|
256
|
Pan QW, Zhong SY, Liu BS, Liu J, Cai R, Wang YG, Liu XY, Qian C. Enhanced sensitivity of hepatocellular carcinoma cells to chemotherapy with a Smac-armed oncolytic adenovirus. Acta Pharmacol Sin 2007; 28:1996-2004. [PMID: 18031615 DOI: 10.1111/j.1745-7254.2007.00672.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AIM The aim of the present study was to further improve the therapeutic effects for human hepatocellular carcinoma (HCC) and reduce the damage in normal cells using a novel chemo-gene-virotherapeutic strategy. METHODS An oncolytic adenoviral vector (ZD55) similar to the typical oncolytic adenovirus ONYX-015, with a deletion of E1B-55K gene, was employed to express the second mitochondria-derived activator of caspases (Smac) protein by constructing a recombinant virus ZD55-Smac. The enhanced cytotoxicity of the combined treatment of ZD55-Smac with cisplatin or 5-fluorouracil (5-FU) was evaluated in several HCC cell lines. Moreover, the negative effects on normal cells have been tested in human normal liver cell lines L-02 and QSG-7701 cell lines by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and apoptotic cell staining. RESULTS According to our observation, ZD55-Smac is superior to ONYX-015 in sensitizing chemotherapy, ZD55-Smac used in conjunction with chemotherapy was found to exhibit obviously enhanced cytotoxicity in HCC cells, yet significantly abolished the negative toxicity in normal cells by utilizing the tumor selective replication vector and reducing the dosage. CONCLUSION This chemo-gene-virotherapeutic (cisplatin or 5-FU+ZD55-Smac) strategy is superior to the conventional chemo-gene or chemo-viro approach.
Collapse
Affiliation(s)
- Qiu-wei Pan
- Xin Yuan Institute of Medicine and Biotechnology, Life Science College, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | | | | | | | | | | | | | | |
Collapse
|
257
|
Nachmias B, Lazar I, Elmalech M, Abed-El-Rahaman I, Asshab Y, Mandelboim O, Perlman R, Ben-Yehuda D. Subcellular localization determines the delicate balance between the anti- and pro-apoptotic activity of Livin. Apoptosis 2007; 12:1129-42. [PMID: 17294084 DOI: 10.1007/s10495-006-0049-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Livin is a member of the Inhibitor of Apoptosis Protein family which inhibits apoptosis induced by a variety of stimuli. We previously identified Livin and demonstrated that following apoptotic stimuli, Livin is cleaved by effector caspases to produce a truncated form with paradoxical pro-apoptotic activity. In the present study, we reveal that while full-length Livin shows diffuse cytoplasmic localization, truncated Livin (tLivin) is found in a peri-nuclear distribution with marked localization to the Golgi apparatus. Using mutation analysis, we identified two domains that are crucial for the pro-apoptotic activity of tLivin: the N-terminal region of tLivin which is exposed by cleavage, and the RING domain. We demonstrate that, of the N-terminal sequence, only the first N-terminal glycine residue dictates the peri-nuclear distribution of tLivin. However, while the perinuclear localization of tLivin is essential, it is not sufficient for tLivin to exert its pro-apoptotic function. Once tLivin is properly localized, an intact RING domain enables its pro-apoptotic function.
Collapse
Affiliation(s)
- Boaz Nachmias
- Department of Hematology, Hadassah - Hebrew University Medical Center, P.O.B. 12000, Jerusalem, 91120, Israel
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Apoptosis-inducing factor is a target for ubiquitination through interaction with XIAP. Mol Cell Biol 2007; 28:237-47. [PMID: 17967870 DOI: 10.1128/mcb.01065-07] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
X-linked inhibitor of apoptosis (XIAP) is an inhibitor of apoptotic cell death that protects cells by caspase-dependent and independent mechanisms. In a screen for molecules that participate with XIAP in regulating cellular activities, we identified apoptosis-inducing factor (AIF) as an XIAP binding protein. Baculoviral IAP repeat 2 of XIAP is sufficient for the XIAP/AIF interaction, which is disrupted by Smac/DIABLO. In healthy cells, mature human AIF lacks only the first 54 amino acids, differing significantly from the apoptotic form, which lacks the first 102 amino-terminal residues. Fluorescence complementation and immunoprecipitation experiments revealed that XIAP interacts with both AIF forms. AIF was found to be a target of XIAP-mediated ubiquitination under both normal and apoptotic conditions, and an E3 ubiquitin ligase-deficient XIAP variant displayed a more robust interaction with AIF. Expression of either XIAP or AIF attenuated both basal and antimycin A-stimulated levels of reactive oxygen species (ROS), and when XIAP and AIF were expressed in combination, a cumulative decrease in ROS was observed. These results identify AIF as a new XIAP binding partner and indicate a role for XIAP in regulating cellular ROS.
Collapse
|
259
|
Lin SC, Huang Y, Lo YC, Lu M, Wu H. Crystal structure of the BIR1 domain of XIAP in two crystal forms. J Mol Biol 2007; 372:847-854. [PMID: 17698078 PMCID: PMC2039701 DOI: 10.1016/j.jmb.2007.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 06/25/2007] [Accepted: 07/10/2007] [Indexed: 10/23/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP) is a potent negative regulator of apoptosis. It also plays a role in BMP signaling, TGF-beta signaling, and copper homeostasis. Previous structural studies have shown that the baculoviral IAP repeat (BIR2 and BIR3) domains of XIAP interact with the IAP-binding-motifs (IBM) in several apoptosis proteins such as Smac and caspase-9 via the conserved IBM-binding groove. Here, we report the crystal structure in two crystal forms of the BIR1 domain of XIAP, which does not possess this IBM-binding groove and cannot interact with Smac or caspase-9. Instead, the BIR1 domain forms a conserved dimer through the region corresponding to the IBM-binding groove. Structural and sequence analyses suggest that this dimerization of BIR1 in XIAP may be conserved in other IAP family members such as cIAP1 and cIAP2 and may be important for the action of XIAP in TGF-beta and BMP signaling and the action of cIAP1 and cIAP2 in TNF receptor signaling.
Collapse
Affiliation(s)
- Su-Chang Lin
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Yihua Huang
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Yu-Chih Lo
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Miao Lu
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Hao Wu
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
260
|
Sung YH, Lee JS, Park SH, Koo J, Lee GM. Influence of co-down-regulation of caspase-3 and caspase-7 by siRNAs on sodium butyrate-induced apoptotic cell death of Chinese hamster ovary cells producing thrombopoietin. Metab Eng 2007; 9:452-64. [PMID: 17892962 DOI: 10.1016/j.ymben.2007.08.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Revised: 08/07/2007] [Accepted: 08/10/2007] [Indexed: 11/28/2022]
Abstract
Previously, the expression of caspase-3 siRNA could not effectively inhibit sodium butyrate (NaBu)-induced apoptotic cell death of recombinant Chinese hamster ovary (rCHO) cells producing human thrombopoietin (hTPO). Caspase-3 siRNA expressing cells appeared to compensate for the lack of caspase-3 by increasing active caspase-7 levels. For the successful inhibition of NaBu-induced apoptosis of rCHO cells, both caspase-3 and caspase-7 were down-regulated using the siRNA expression vector system. Co-down-regulation of caspase-3 and caspase-7 increased cell viability and extended culture longevity in serum-free culture in the presence or absence of 1mM NaBu addition. In the cultures with 1mM NaBu addition, the maximum hTPO concentration in rCHO cells with down-regulation of both caspases was approximately 55% higher than that in rCHO cells without down-regulation of caspases and approximately 16% higher than rCHO cells with down-regulation of only caspase-3. However, in the culture with 3mM NaBu, this strategy could not dramatically enhance the culture longevity and hTPO production, compared to Bcl-2 overexpression. The different result in hTPO production between down-regulation of caspases and Bcl-2 overexpression may be because the down-regulation of caspase-3 and caspase-7, unlike Bcl-2 overexpression, could not maintain mitochondrial membrane potential in the presence of 3mM NaBu. Taken together, co-down-regulation of caspase-3 and caspase-7 is effective in regard to extension of culture longevity and enhancement of hTPO production in a serum-free culture in the presence or absence of 1mM NaBu addition.
Collapse
Affiliation(s)
- Yun Hee Sung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 373-1 Kusong-Dong, Yusong-Gu, Daejon 305-701, Republic of Korea
| | | | | | | | | |
Collapse
|
261
|
Gao Z, Tian Y, Wang J, Yin Q, Wu H, Li YM, Jiang X. A dimeric Smac/diablo peptide directly relieves caspase-3 inhibition by XIAP. Dynamic and cooperative regulation of XIAP by Smac/Diablo. J Biol Chem 2007; 282:30718-27. [PMID: 17724022 PMCID: PMC3202417 DOI: 10.1074/jbc.m705258200] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Caspase activation, the executing event of apoptosis, is under deliberate regulation. IAP proteins inhibit caspase activity, whereas Smac/Diablo antagonizes IAP. XIAP, a ubiquitous IAP, can inhibit both caspase-9, the initiator caspase of the mitochondrial apoptotic pathway, and the downstream effector caspases, caspase-3 and caspase-7. Smac neutralizes XIAP inhibition of caspase-9 by competing for binding of the BIR3 domain of XIAP with caspase-9, whereas how Smac liberates effector caspases from XIAP inhibition is not clear. It is generally believed that binding of Smac with IAP generates a steric hindrance that prevents XIAP from inhibiting effector caspases, and therefore small molecule mimics of Smac are not able to reverse inhibition of the effector caspases. Surprisingly, we show here that binding of a dimeric Smac N-terminal peptide with the BIR2 domain of XIAP effectively antagonizes inhibition of caspase-3 by XIAP. Further, we defined the dynamic and cooperative interaction of Smac with XIAP: binding of Smac with the BIR3 domain anchors the subsequent binding of Smac with the BIR2 domain, which in turn attenuates the caspase-3 inhibitory function of XIAP. We also show that XIAP homotrimerizes via its C-terminal Ring domain, making its inhibitory activity toward caspase-3 more susceptible to Smac.
Collapse
Affiliation(s)
- Zhonghua Gao
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
| | - Yuan Tian
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
| | - Junru Wang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Qian Yin
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - Hao Wu
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
- Graduate School of Biomedical Sciences, Weill Medical College of Cornell University, New York, NY 10021
| |
Collapse
|
262
|
Mufti AR, Burstein E, Duckett CS. XIAP: cell death regulation meets copper homeostasis. Arch Biochem Biophys 2007; 463:168-74. [PMID: 17382285 PMCID: PMC1986780 DOI: 10.1016/j.abb.2007.01.033] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 01/23/2007] [Accepted: 01/25/2007] [Indexed: 12/11/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP), traditionally known as an anti-apoptotic protein, has recently been shown to be involved in copper homeostasis. XIAP promotes the ubiquitination and degradation of COMMD1, a protein that promotes the efflux of copper from the cell. Through its effects on COMMD1, XIAP can regulate copper export from the cell and potentially represents an additional intracellular sensor for copper levels. XIAP binds copper directly and undergoes a substantial conformational change in the copper-bound state. This in turn destabilizes XIAP, resulting in lowered steady-state levels of the protein. Furthermore, copper-bound XIAP is unable to inhibit caspases and cells that express this form of the protein exhibit increased rates of cell death in response to apoptotic stimuli. These events take place in the setting of excess intracellular copper accumulation as seen in copper toxicosis disorders such as Wilson's disease and establish a new relationship between copper levels and the regulation of cell death via XIAP. These findings raise important questions about the role of XIAP in the development of copper toxicosis disorders and may point to XIAP as a potential therapeutic target in these disease states.
Collapse
Affiliation(s)
- Arjmand R Mufti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | |
Collapse
|
263
|
Lu M, Lin SC, Huang Y, Kang YJ, Rich R, Lo YC, Myszka D, Han J, Wu H. XIAP induces NF-kappaB activation via the BIR1/TAB1 interaction and BIR1 dimerization. Mol Cell 2007; 26:689-702. [PMID: 17560374 PMCID: PMC1991276 DOI: 10.1016/j.molcel.2007.05.006] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 04/03/2007] [Accepted: 05/07/2007] [Indexed: 11/20/2022]
Abstract
In addition to caspase inhibition, X-linked inhibitor of apoptosis (XIAP) induces NF-kappaB and MAP kinase activation during TGF-b and BMP receptor signaling and upon overexpression. Here we show that the BIR1 domain of XIAP, which has no previously ascribed function, directly interacts with TAB1 to induce NF-kappaB activation. TAB1 is an upstream adaptor for the activation of the kinase TAK1, which in turn couples to the NF-kappaB pathway. We report the crystal structures of BIR1, TAB1, and the BIR1/TAB1 complex. The BIR1/TAB1 structure reveals a striking butterfly-shaped dimer and the detailed interaction between BIR1 and TAB1. Structure-based mutagenesis and knockdown of TAB1 show unambiguously that the BIR1/TAB1 interaction is crucial for XIAP-induced TAK1 and NF-kappaB activation. We show that although not interacting with BIR1, Smac, the antagonist for caspase inhibition by XIAP, also inhibits the XIAP/TAB1 interaction. Disruption of BIR1 dimerization abolishes XIAP-mediated NF-kappaB activation, implicating a proximity-induced mechanism for TAK1 activation.
Collapse
Affiliation(s)
- Miao Lu
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - Su-Chang Lin
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - Yihua Huang
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - Young Jun Kang
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Rebecca Rich
- Center for Biomolecular Interaction Analysis, School of Medicine, University of Utah, Salt Lake City, Utah 84132
| | - Yu-Chih Lo
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| | - David Myszka
- Center for Biomolecular Interaction Analysis, School of Medicine, University of Utah, Salt Lake City, Utah 84132
| | - Jiahuai Han
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Hao Wu
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021
| |
Collapse
|
264
|
Kurakin A, Bredesen DE. An unconventional IAP-binding motif revealed by target-assisted iterative screening (TAIS) of the BIR3-cIAP1 domain. J Mol Recognit 2007; 20:39-50. [PMID: 17094177 PMCID: PMC1847359 DOI: 10.1002/jmr.809] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Target-assisted iterative screening (TAIS) has been applied to a random phage-displayed peptide library in a search for novel ligands of the third baculovirus IAP ('inhibitors of apoptosis') repeat (BIR) domain of cIAP1. The peptides selected in the screen fall into two distinct specificity groups, one that conforms to a known IAP-binding motif (IBM) and another one that reveals a novel BIR domain interacting motif, NH(2)-SR(V/P)W. The biochemical profiling of selected sequences with synthetic peptides, which included alanine scanning and N- and C-terminal truncations as well as competition with the Smac peptide, suggests a major energetic contribution of tryptophan at the +4 position of peptide ligands to binding and identifies the latter together with the respective pocket on the BIR domain surface as a 'hot spot' of the interaction. A peptide featuring the novel motif selectively binds the full-length cIAP1 protein in cell lysates. A 'two-pocket' model of BIR domain recognition mechanism is proposed as the basis of differential BIR domain interactions with different IBMs.
Collapse
Affiliation(s)
- Alexei Kurakin
- The Buck Institute for Age Research, 8001 Redwood Blvd., Novato, CA 94945, USA.
| | | |
Collapse
|
265
|
Abstract
Defects in the core machinery of the apoptosis pathway contribute to chemoresistance and poor outcomes in patients with acute leukemia. To overcome these defects, novel molecules that target key proteins in the apoptosis pathway are being developed. This review highlights compounds that target the mitochondrial, death receptor, and convergence pathways of caspase activation that are being developed for the treatment of acute leukemia.
Collapse
Affiliation(s)
- Aaron D Schimmer
- Princess Margaret Hospital, Room 9-516, 610 University Avenue, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|
266
|
Abstract
We have developed a two-component system involving reconstituted caspase (recCaspase) for selective and/or conditional ablation of targeted cells. Caspases, the executioners of programmed cell death, are normally synthesized as inactive zymogens and are activated by proteolytic processing of their subunits. We show here, using two different caspases, Caenorhabditis elegans CED-3 and human Caspase-3, that coexpression of the subunits generates constitutively active caspase activity that leads to cell death. This recCaspase activity, however, occurred only when the subunits associated through binding of linked antiparallel leucine-zipper domains. We exploited the dual-component nature of recCaspases by expressing the individual subunits from combinations of promoters either to target selectively the subset of cells for apoptosis or induce cell death in specific cells at specific times during development. The high degree of target specificity and tight regulation of induction of recCaspase would be advantageous in creating animal models that are ablated for specific cells and in other targeted cell killings.
Collapse
Affiliation(s)
- Dattananda S. Chelur
- Department of Biological Sciences, Columbia University, 1012 Fairchild Center, New York, NY 10027
- To whom correspondence may be addressed. E-mail:
or
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, 1012 Fairchild Center, New York, NY 10027
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
267
|
Zhang C, Xie L, Cheng H, Wang Y. TRAF3 interacts with Smac/DIABLO and enhances the proapoptotic effect of Smac/DIABLO in cytoplasm. Acta Biochim Biophys Sin (Shanghai) 2007; 39:108-16. [PMID: 17277885 DOI: 10.1111/j.1745-7270.2007.00259.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Smac/DIABLO (second mitochondria-derived activator of caspase/direct IAP-binding protein with low PI) is a 29 kDa mitochondrial precursor protein, which is proteolytically processed in mitochondria into a 23 kDa mature protein. It is released from the mitochondrial intermembrane space to cytosol after an apoptotic trigger. Smac/DIABLO acts as a dimer and it contributes to caspase activation by sequestering the inhibitor of apoptosis proteins (IAPs). In order to further investigate the mechanism of Smac/DIABLO action, we used the mature form of Smac/DIABLO as a bait and screened proteins that interact with mature Smac/DIABLO in human liver cDNA library using the yeast two-hybrid system. Forty-two colonies were obtained after 5.8x10(6) colonies were screened by nutrition limitation and X-galactosidase assay. After DNA sequence analysis and homology retrieval, one of the candidate proteins was identified as TRAF domain of the TNF receptor associated factor 3 (TRAF3). The interaction site between TRAF3 and Smac/DIABLO was identified by beta-galactosidase test. The interaction between TRAF3 and Smac/DIABLO via TRAF domain was identified in vivo by co-immunoprecipitation in HepG2 cells, and the direct interaction between TRAF3 and Smac/DIABLO in vitro was identified by GST-pull down assay. Co-expression of TRAF3 and mature Smac/DIABLO in 293 cells could enhance the Smac/DIABLO-mediated apoptosis. These results suggested that TRAF3 interacted with Smac/DIABLO via TRAF domain, leading to an increased proapoptotic effect of Smac/DIABLO in cytoplasm.
Collapse
Affiliation(s)
- Cuili Zhang
- Clinical Laboratory Sichuan Corps Hospital of The People's Armed Police Forces, Leshan 614000, China
| | | | | | | |
Collapse
|
268
|
Liou JY, Matijevic-Aleksic N, Lee S, Wu KK. Prostacyclin inhibits endothelial cell XIAP ubiquitination and degradation. J Cell Physiol 2007; 212:840-8. [PMID: 17450518 DOI: 10.1002/jcp.21082] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To understand the role of prostacyclin (PGI(2)) in protecting endothelial cells from apoptosis, we evaluated the effects of carbaprostacyclin (cPGI(2)) on H(2)O(2)-induced human umbilical vein endothelial cell (HUVEC) apoptosis. cPGI(2) suppressed H(2)O(2)-induced annexin V-positive cells in a concentration- and time-dependent manner. Pre-treatment of HUVEC with 50 microM cPGI(2) for 4 h produced the maximal anti-apoptotic effect. Authentic PGI(2) generated by adenoviral transfer of PGI(2) synthetic genes exerted a similar protective effect. cPGI(2) inhibited Smac/DIABLO release from mitochondria, caspase 3 activation, focal adhesion protein degradation, and cell detachment. cPGI(2) selectively protected X-linked inhibitor of apoptosis protein (X-linked IAP, XIAP) from H(2)O(2)-induced ubiquitination, and preserved XIAP protein levels. PD-98059 but not H-89 abrogated the protective action of cPGI(2). cPGI(2) increased ERK phosphorylation which was blocked by PD-98059. HUVEC stably transfected with dominant negative Ras abrogated XIAP preservation by cPGI(2) while constitutive active Ras increased ERK phosphorylation and protected XIAP from degradation. Our results demonstrate for the first time that PGI(2) inhibits XIAP ubiquitination and degradation via the Ras/MEK-1/ERK signaling pathway. Preservation of XIAP proteins represents a key mechanism by which PGI(2) protects endothelial cells from oxidant-induced apoptosis.
Collapse
Affiliation(s)
- Jun-Yang Liou
- University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
269
|
Checinska A, Hoogeland BSJ, Rodriguez JA, Giaccone G, Kruyt FAE. Role of XIAP in inhibiting cisplatin-induced caspase activation in non-small cell lung cancer cells: a small molecule Smac mimic sensitizes for chemotherapy-induced apoptosis by enhancing caspase-3 activation. Exp Cell Res 2006; 313:1215-24. [PMID: 17291493 DOI: 10.1016/j.yexcr.2006.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 11/18/2006] [Accepted: 12/20/2006] [Indexed: 01/10/2023]
Abstract
X-linked IAP (XIAP) suppresses apoptosis by binding to initiator caspase-9 and effector caspases-3 and -7. Smac/DIABLO that is released from mitochondria during apoptosis can relieve its inhibitory activity. Here we investigated the role of XIAP in the previously found obstruction of chemotherapy-induced caspase-9 activation in non-small cell lung cancer (NSCLC) cells. Endogenously expressed XIAP bound active forms of both caspase-9 and caspase-3. However, downregulation of XIAP using shRNA or disruption of XIAP/caspase-9 interaction using a small molecule Smac mimic were unable to significantly induce caspase-9 activity, indicating that despite a strong binding potential of XIAP to caspase-9 it is not a major determinant in blocking caspase-9 in NSCLC cells. Although unable to revert caspase-9 blockage, the Smac mimic was able to enhance cisplatin-induced apoptosis, which was accompanied by increased caspase-3 activity. Additionally, a more detailed analysis of caspase activation in response to cisplatin indicated a reverse order of activation, whereby caspase-3 cleaved caspase-9 yielding an inactive form. Our findings indicate that the use of small molecule Smac mimic, when combined with an apoptotic trigger, may have therapeutic potential for the treatment of NSCLC.
Collapse
Affiliation(s)
- Agnieszka Checinska
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, CCA 2.44 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
270
|
Feeney B, Pop C, Swartz P, Mattos C, Clark AC. Role of loop bundle hydrogen bonds in the maturation and activity of (Pro)caspase-3. Biochemistry 2006; 45:13249-63. [PMID: 17073446 PMCID: PMC3119718 DOI: 10.1021/bi0611964] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During maturation, procaspase-3 is cleaved at D175, which resides in a linker that connects the large and small subunits. The intersubunit linker also connects two active site loops that rearrange following cleavage and, in part, form the so-called loop bundle. As a result of chain cleavage, new hydrogen bonds and van der Waals contacts form among three active site loops. The new interactions are predicted to stabilize the active site. One unresolved issue is the extent to which the loop bundle residues also stabilize the procaspase active site. We examined the effects of replacing four loop bundle residues (E167, D169, E173, and Y203) on the biochemical and structural properties of the (pro)caspase. We show that replacing the residues affects the activity of the procaspase as well as the mature caspase, with D169A and E167A replacements having the largest effects. Replacement of D169 prevents caspase-3 autoactivation, and its cleavage at D175 no longer leads to an active enzyme. In addition, the E173A mutation, when coupled to a second mutation in the procaspase, D175A, may alter the substrate specificity of the procaspase. The mutations affected the active site environment as assessed by changes in fluorescence emission, accessibility to quencher, and cleavage by either trypsin or V8 proteases. High-resolution X-ray crystallographic structures of E167A, D173A, and Y203F caspases show that changes in the active site environment may be due to the increased flexibility of several residues in the N-terminus of the small subunit. Overall, the results show that these residues are important for stabilizing the procaspase active site as well as that of the mature caspase.
Collapse
Affiliation(s)
- Brett Feeney
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27695
| | | | - Paul Swartz
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27695
| | - Carla Mattos
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27695
| | - A. Clay Clark
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
271
|
Eckelman BP, Salvesen GS, Scott FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep 2006; 7:988-94. [PMID: 17016456 PMCID: PMC1618369 DOI: 10.1038/sj.embor.7400795] [Citation(s) in RCA: 605] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 07/31/2006] [Indexed: 12/13/2022] Open
Abstract
Several of the inhibitor of apoptosis protein (IAP) family members regulate apoptosis in response to various cellular assaults. Some members are also involved in cell signalling, mitosis and targeting proteins to the ubiquitin-proteasome degradation machinery. The most intensively studied family member, X-linked IAP (XIAP), is a potent inhibitor of caspase activity; hence, it is generally assumed that direct caspase inhibition is an important conserved function of most members of the family. Biochemical and structural studies have precisely mapped the elements of XIAP required for caspase inhibition. Intriguingly, these elements are not conserved among IAPs. Here, we review current knowledge of the caspase-inhibitory potential of the human IAPs and show that XIAP is probably the only bona fide caspase inhibitor, suggesting that the other family members never gained the ability to directly inhibit caspase activity.
Collapse
Affiliation(s)
- Brendan P Eckelman
- Program in Apoptosis and Cell Death Research, Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, California 92037, USA
| | - Guy S Salvesen
- Program in Apoptosis and Cell Death Research, Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, California 92037, USA
| | - Fiona L Scott
- Program in Apoptosis and Cell Death Research, Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
272
|
Qi R, Gu J, Zhang Z, Yang K, Li B, Fan J, Wang C, He Z, Qiao L, Lin Z, Liu XY. Potent antitumor efficacy of XAF1 delivered by conditionally replicative adenovirus vector via caspase-independent apoptosis. Cancer Gene Ther 2006; 14:82-90. [PMID: 17008933 DOI: 10.1038/sj.cgt.7700992] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
XAF1 is a newly identified tumor-suppressor gene that can antagonize XIAP and sensitize cells to other cell death triggers. In this study, we utilized ZD55, a conditionally replicative adenovirus (CRAd) similar to ONYX-015 as the vector to transfer XAF1 into the tumor cells to evaluate its antitumor efficacy in vitro and in vivo. Potent and specific cytopathic effect (CPE) was observed upon infection with ZD55-XAF1 in tumor cell lines. Importantly, ZD55-XAF1 exhibited a superior suppression of tumor growth in an animal model of colorectal carcinoma in nude mice compared with Ad-XAF1 (E1-deleted replication-defective viral) and ONYX-015. Complete eradication of the established tumors was observed in four of eight mice. Our data also showed that infection with ZD55-XAF1 resulted in caspase-independent apoptosis. Although caspase-3, poly(ADP-ribose) polymerase were mildly activated in response to ZD55-XAF1 infection, pretreatment with pan-caspase inhibitor hardly influence its apoptosis-inducing activity. In summary, our study strongly suggested that ZD55-XAF1 could serve as an effective gene-virotherapy strategy and has highly potential against human cancers.
Collapse
Affiliation(s)
- R Qi
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
273
|
N/A, 刘 芝, 张 林. N/A. Shijie Huaren Xiaohua Zazhi 2006; 14:2626-2631. [DOI: 10.11569/wcjd.v14.i26.2626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
274
|
Abu-Zant A, Jones S, Asare R, Suttles J, Price C, Graham J, Kwaik YA. Anti-apoptotic signalling by the Dot/Icm secretion system of L. pneumophila. Cell Microbiol 2006; 9:246-64. [PMID: 16911566 DOI: 10.1111/j.1462-5822.2006.00785.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Dot/Icm type IV secretion system of Legionella pneumophila triggers robust activation of caspase-3 during early and exponential stages of proliferation within human macrophages, but apoptosis is delayed till late stages of infection, which is novel. As caspase-3 is the executioner of the cell, we tested the hypothesis that L. pneumophila triggers anti-apoptotic signalling within the infected human macrophages to halt caspase-3 from dismantling the cells. Here we show that during early and exponential replication, L. pneumophila-infected human monocyte-derived macrophages (hMDMs) exhibit a remarkable resistance to induction of apoptosis, in a Dot/Icm-dependent manner. Microarray analyses and real-time PCR reveal that during exponential intracellular replication, L. pneumophila triggers upregulation of 12 anti-apoptotic genes that are linked to activation of the nuclear transcription factor kappa-B (NF-kappaB). Our data show that L. pneumophila induces a Dot/Icm-dependent sustained nuclear translocation of the p50 and p65 subunits of NF-kappaB during exponential intracellular replication. Bacterial entry is essential both for the anti-apoptotic phenotype of infected hMDMs and for nuclear translocation of the p65. Using p65-/- and IKKalpha-/- beta-/- double knockout mouse embryonic fibroblast cell lines, we show that nuclear translocation of NF-kappaB is required for the resistance of L. pneumophila-infected cells to apoptosis-inducing agents. In addition, the L. pneumophila-induced nuclear translocation of NF-kappaB requires the activity of IKKalpha and/or IKKbeta. We conclude that although the Dot/Icm secretion system of L. pneumophila elicits an early robust activation of caspase-3 in human macrophages, it triggers a strong anti-apoptotic signalling cascade mediated, at least in part by NF-kappaB, which renders the cells refractory to external potent apoptotic stimuli.
Collapse
Affiliation(s)
- Alaeddin Abu-Zant
- Department of Microbiology, University of Louisville Collage of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
275
|
Abstract
Accumulating evidence indicates that there is a critical role of the ubiquitin/proteasome pathway in the regulation of apoptosis. Among the important molecules that couple these two fundamental cellular activities are members of the inhibitor of apoptosis (IAP) protein family. In addition to their well-studied ability to directly bind and inhibit caspases, many IAPs contain RING domains that are necessary and sufficient to cause ubiquitylation and subsequent proteasome-mediated proteolysis. This review summarizes recent findings about the ubiquitin protein ligase activity of IAPs, and considers possible mechanisms for substrate selectivity.
Collapse
Affiliation(s)
- Ting Ni
- College of Life Science, Sichuan University, Sichuan Province, P. R. China
| | | | | |
Collapse
|
276
|
Li T, Lu C, Xia Z, Xiao B, Luo Y. Inhibition of caspase-8 attenuates neuronal death induced by limbic seizures in a cytochrome c-dependent and Smac/DIABLO-independent way. Brain Res 2006; 1098:204-11. [PMID: 16774749 DOI: 10.1016/j.brainres.2006.04.131] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 04/14/2006] [Accepted: 04/19/2006] [Indexed: 11/18/2022]
Abstract
There is increasing evidence that neuronal cell death induced by seizures occurs via extrinsic (death receptors) and intrinsic (mitochondria) pathways. Caspase-8 cleaves Bid, which releases cytochrome c, bridging the "extrinsic" and "intrinsic" pathways. Cleavage of Bid may amplify caspase-8-induced neuronal cell death following seizures. In the present study, we explored the effect of an inhibitor of caspase-8 (z-IETD-fmk) on the release of Smac/DIABLO and cytochrome c from mitochondria. Rats received intra-amygdaloid injection of kainic acid (KA) to induce seizures for 1 h. The seizures were then terminated by diazepam (30 mg/kg). The damaged and surviving neurons in hippocampus were observed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and cresyl violet staining, the expression of caspase-8, Bid, XIAP, caspase-9, cytochrome c and Smac/DIABLO were detected with immunofluorescence and Western blot. The cleavage of caspase-8 and Bid increased at 0 h, cytosolic fraction of cytochrome c and Smac/DIABLO increased by 2 h, cleavage of caspase-9 was detected by 4 h, TUNEL-positive neurons appeared at 8 h and reached a maximum at 24 h following administration of diazepam in the ipsilateral CA3 subfield of hippocampus. Inhibition of caspase-8 significantly decreased neuronal cell death, accompanied by reduction of t-Bid, cleaved caspase-9 and cytosol cytochrome c. Smac/DIABLO from mitochondria was not affected. These results suggest that seizures can lead the translocation of cytochrome c into the cytosol, and the activation of caspase-8 occurs upstream the mitochondria release of cytochrome c and Smac/DIABLO. Inhibition of caspase-8 attenuated neuronal cell death following seizures by decreasing mitochondria release of cytochrome c but not Smac/DIABLO.
Collapse
Affiliation(s)
- Tianfu Li
- Institute of Neurology, Huashan Hospital, University of Fudan, 200040 Shanghai, China
| | | | | | | | | |
Collapse
|
277
|
Siskind LJ, Kolesnick RN, Colombini M. Ceramide forms channels in mitochondrial outer membranes at physiologically relevant concentrations. Mitochondrion 2006; 6:118-25. [PMID: 16713754 PMCID: PMC2246045 DOI: 10.1016/j.mito.2006.03.002] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Revised: 03/06/2006] [Accepted: 03/07/2006] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that the ability of ceramides to induce apoptosis is due to a direct action on mitochondria. Mitochondria are known to contain enzymes responsible for ceramide synthesis and hydrolysis and mitochondrial ceramide levels have been shown to be elevated prior to the mitochondrial phase of apoptosis. Ceramides have been reported to induce the release of intermembrane space proteins from mitochondria, which has been linked to their ability to form large channels in membranes. The aim of this study was to determine if the membrane concentration of ceramide required for the formation of protein permeable channels is within the range that is present in mitochondria during the induction phase of apoptosis. Only a very small percentage of the ceramide actually inserts into the mitochondrial membranes. The permeability of the mitochondrial outer membrane correlates directly with the level of ceramide in the membrane. Importantly, the concentration of ceramide at which significant channel formation occurs is consistent with the level of mitochondrial ceramide that occurs during the induction phase of apoptosis (4 pmol ceramide/nanomole phospholipid). Similar results were obtained with short- and long-chain ceramide. Ceramide channel formation is specific to mitochondrial membranes in that no channel formation occurs in the plasma membranes of erythrocytes even at concentrations 20 times higher than those required for channel formation in mitochondrial outer membranes. Thus, ceramide channels are good candidates for the pathway by which proapoptotic proteins are released from mitochondria during the induction phase of apoptosis.
Collapse
Affiliation(s)
- Leah J. Siskind
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Richard N. Kolesnick
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Marco Colombini
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
278
|
Salako MA, Carter MJ, Kass GEN. Coxsackievirus protein 2BC blocks host cell apoptosis by inhibiting caspase-3. J Biol Chem 2006; 281:16296-304. [PMID: 16608851 DOI: 10.1074/jbc.m510662200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Virus infection may induce host cell death by apoptosis, but some DNA viruses are capable of preventing this process. RNA viruses were thought not to display anti-apoptotic activities, as their spread appears to benefit from a rapid induction of cell death. Here, we report an antiapoptotic activity in the Picornavirus Coxsackievirus B4 (CVB4). CVB4 infection of HeLa cells induced negligible apoptosis over a period of 10 h. However, infected cells developed resistance to drug-induced apoptosis using staurosporine and actinomycin D and to death receptor-induced apoptosis using tumor necrosis factor-related apoptosis-inducing ligand. Despite this resistance, the apoptotic machinery was nonetheless fully activated in these drug-treated infected cells because the levels of pro-caspase-3 processing to its active form were similar to control cells. However, the DEVDase (Asp-Glu-Val-Asp protease) activity of the processed caspase was significantly inhibited in the virus-infected staurosporine-treated cells compared with drug treatment alone. Likewise, extracts of CVB4-infected cells suppressed recombinant caspase-3 activity in vitro. Immunoprecipitation of activated caspase-3 from radiolabeled virus-infected cells revealed the co-precipitation of a 48-kDa protein that was tentatively identified as viral protein 2BC. Recombinant caspase-3 was found to co-precipitate with virus protein 2BC. Finally, when protein 2BC was expressed in HeLa cells, both staurosporine-induced apoptosis and in vitro caspase-3 DEVDase activity were significantly reduced. Taken together these data imply that CVB4 infection suppresses apoptosis through virus protein 2BC associating with caspase-3 and inhibiting its function. Thus, 2BC is the first reported RNA virus inhibitor of apoptosis protein.
Collapse
Affiliation(s)
- Michael A Salako
- School of Biomedical and Molecular Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | | | | |
Collapse
|
279
|
|
280
|
Li Y, Jian Z, Xia K, Li X, Lv X, Pei H, Chen Z, Li J. XIAP is related to the chemoresistance and inhibited its expression by RNA interference sensitize pancreatic carcinoma cells to chemotherapeutics. Pancreas 2006; 32:288-96. [PMID: 16628085 DOI: 10.1097/01.mpa.0000218314.67111.fb] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To explore the exact role does the x-linked inhibitor of apoptosis (XIAP) play in chemoresistance of pancreatic carcinoma cell and the cell sensitivity to chemotherapeutic drugs changed after XIAP is inhibited by RNA interference (RNAi). METHODS Pancreatic carcinoma cell line SW1990 was exposed to 5-fluorouracil (5-fu) with the concentrations of 1.0 and 10 mug/mL to increase the expression of XIAP. Then 4 RNAi plasmid vectors against XIAP were designed and constructed, then transfected into SW1990. Repressive effect was evaluated by reverse transcriptase polymerase chain reaction and Western blot; 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometry were performed to determine cell sensitivity to 5-fu and gemcitabine; furthermore, apoptosis is confirmed by Hoechst 33258 stain. RESULTS XIAP of SW1990 can be up-regulated with the chemoresistance increasing 1.5- and 4-fold after 10 and 30 days induced by 5-fu. Two of the 4 vectors can inhibit the expression of XIAP protein more than 60%. The cells apoptosis index induced by 5-fu and gemcitabine increased greatly after XIAP is inhibited by the RNAi plasmid vectors. CONCLUSIONS XIAP is one of the most important factors in the pancreatic carcinoma chemoresistance, and inhibition of XIAP in pancreatic carcinoma can enhance the cancer sensitivity to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Yixiong Li
- Department of General Surgery, Xiangya Hospital, Hunan, China.
| | | | | | | | | | | | | | | |
Collapse
|
281
|
Lamb HK, Mee C, Xu W, Liu L, Blond S, Cooper A, Charles IG, Hawkins AR. The affinity of a major Ca2+ binding site on GRP78 is differentially enhanced by ADP and ATP. J Biol Chem 2006; 281:8796-805. [PMID: 16418174 DOI: 10.1074/jbc.m503964200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
GRP78 is a major protein regulated by the mammalian endoplasmic reticulum stress response, and up-regulation has been shown to be important in protecting cells from challenge with cytotoxic agents. GRP78 has ATPase activity, acts as a chaperone, and interacts specifically with other proteins, such as caspases, as part of a mechanism regulating apoptosis. GRP78 is also reported to have a possible role as a Ca2+ storage protein. In order to understand the potential biological effects of Ca2+ and ATP/ADP binding on the biology of GRP78, we have determined its ligand binding properties. We show here for the first time that GRP78 can bind Ca2+, ATP, and ADP, each with a 1:1 stoichiometry, and that the binding of cation and nucleotide is cooperative. These observations do not support the hypothesis that GRP78 is a dynamic Ca2+ storage protein. Furthermore, we demonstrate that whereas Mg2+ enhances GRP78 binding to ADP and ATP to the same extent, Ca2+ shows a differential enhancement. In the presence of Ca2+, the KD for ATP is lowered approximately 11-fold, and the KD for ADP is lowered around 930-fold. The KD for Ca2+ is lowered approximately 40-fold in the presence of ATP and around 880-fold with ADP. These findings may explain the biological requirement for a nucleotide exchange factor to remove ADP from GRP78. Taken together, our data suggest that the Ca2+-binding property of GRP78 may be part of a signal transduction pathway that modulates complex interactions between GRP78, ATP/ADP, secretory proteins, and caspases, and this ultimately has important consequences for cell viability.
Collapse
Affiliation(s)
- Heather K Lamb
- Institute of Cell and Molecular Biosciences, Medical School, University of Newcastle upon Tyne, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
282
|
Schimmer AD, Dalili S, Batey RA, Riedl SJ. Targeting XIAP for the treatment of malignancy. Cell Death Differ 2006; 13:179-88. [PMID: 16322751 DOI: 10.1038/sj.cdd.4401826] [Citation(s) in RCA: 223] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is a member of the inhibitor of apoptosis proteins family of caspase inhibitors that selectively binds and inhibits caspases-3, -7 and -9, but not caspase-8. As such, XIAP blocks a substantial portion of the apoptosis pathway and is an attractive target for novel therapeutic agents for the treatment of malignancy. Antisense oligonucleotides directed against XIAP are effective in vitro and are currently being evaluated in clinical trials. Small molecule XIAP inhibitors that target the baculovirus IAP repeat (BIR) 2 or BIR 3 domain are in preclinical development and are advancing toward the clinic. This review will discuss the progress being made in developing antisense and small-molecule XIAP inhibitors.
Collapse
Affiliation(s)
- A D Schimmer
- The Ontario Cancer Institute, Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
283
|
Sharma SK, Straub C, Zawel L. Development of Peptidomimetics Targeting IAPs. Int J Pept Res Ther 2006; 12:21-32. [PMID: 19617919 PMCID: PMC2710984 DOI: 10.1007/s10989-005-9003-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2005] [Indexed: 11/21/2022]
Abstract
Inhibitor of apoptosis proteins (IAPs) such as XIAP subvert apoptosis by binding and inhibiting caspases. Because occupation of the XIAP BIR3 peptide binding pocket by Smac abolishes the XIAP–caspase 9 interaction, it is a proapoptotic event of great therapeutic interest. An assay for pocket binding was developed based on the displacement of Smac 7-mer from BIR3. Through the physical and biochemical analysis of a variety of peptides, we have determined the minimum sequence required for inhibition of the Smac–BIR3 interaction and detailed the dimensions and topology of the BIR3 peptide binding pocket. This work describes the structure–activity relationship (SAR) for peptide inhibitors of Smac-IAP binding.
Collapse
|
284
|
Mufti AR, Burstein E, Csomos RA, Graf PCF, Wilkinson JC, Dick RD, Challa M, Son JK, Bratton SB, Su GL, Brewer GJ, Jakob U, Duckett CS. XIAP Is a Copper Binding Protein Deregulated in Wilson's Disease and Other Copper Toxicosis Disorders. Mol Cell 2006; 21:775-85. [PMID: 16543147 DOI: 10.1016/j.molcel.2006.01.033] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 11/18/2005] [Accepted: 01/30/2006] [Indexed: 10/24/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP), known primarily for its caspase inhibitory properties, has recently been shown to interact with and regulate the levels of COMMD1, a protein associated with a form of canine copper toxicosis. Here, we describe a role for XIAP in copper metabolism. We find that XIAP levels are greatly reduced by intracellular copper accumulation in Wilson's disease and other copper toxicosis disorders and in cells cultured under high copper conditions. Elevated copper levels result in a profound, reversible conformational change in XIAP due to the direct binding of copper to XIAP, which accelerates its degradation and significantly decreases its ability to inhibit caspase-3. This results in a lowering of the apoptotic threshold, sensitizing the cell to apoptosis. These data provide an unsuspected link between copper homeostasis and the regulation of cell death through XIAP and may contribute to the pathophysiology of copper toxicosis disorders.
Collapse
Affiliation(s)
- Arjmand R Mufti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
285
|
Abstract
Death receptor signaling is initiated by the assembly of the death-inducing signaling complex, which culminates in the activation of the initiator caspase, either caspase-8 or caspase-10. A family of viral and cellular proteins, known as FLIP, plays an essential role in the regulation of death receptor signaling. Viral FLIP (v-FLIP) and short cellular FLIP (c-FLIPS) inhibit apoptosis by interfering with death receptor signaling. The structure and mechanisms of v-FLIP and c-FLIPS remain largely unknown. Here we report a high resolution crystal structure of MC159, a v-FLIP derived from the molluscum contagiosum virus, which is a member of the human poxvirus family. Unexpectedly, the two tandem death effector domains (DEDs) of MC159 rigidly associate with each other through a hydrophobic interface. Structure-based sequence analysis suggests that this interface is conserved in the tandem DEDs from other v-FLIP, c-FLIPS, and caspase-8 and -10. Strikingly, the overall packing arrangement between the two DEDs of MC159 resembles that between the caspase recruitment domains of Apaf-1 and caspase-9. In addition, each DED of MC159 contains a highly conserved binding motif on the surface, to which loss-of-function mutations in MC159 map. These observations, in conjunction with published evidence, reveal significant insights into the function of v-FLIP and suggest a mechanism by which v-FLIP and c-FLIPS inhibit death receptor signaling.
Collapse
Affiliation(s)
- Feng-Yen Li
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Princeton, New Jersey 08544, USA
| | | | | | | |
Collapse
|
286
|
Ravi R, Fuchs EJ, Jain A, Pham V, Yoshimura K, Prouser T, Jalla S, Zhou X, Garrett-Mayer E, Kaufmann SH, Schulick RD, Pardoll DM, Bedi A. Resistance of Cancers to Immunologic Cytotoxicity and Adoptive Immunotherapy via X-Linked Inhibitor of Apoptosis Protein Expression and Coexisting Defects in Mitochondrial Death Signaling. Cancer Res 2006; 66:1730-9. [PMID: 16452233 DOI: 10.1158/0008-5472.can-05-3377] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ability of cancers to evade immune surveillance and resist immunotherapy raises a fundamental question of how tumor cells survive in the presence of a competent immune system. Studies to address this question have primarily focused on mechanisms by which tumor cells avoid recognition by or induce tolerance in the immune system. However, little is known about whether cancer cells also acquire an intrinsic ability to resist killing by immune effectors. We find that cancer cells enhance their ability to withstand an attack by cytotoxic immune effector cells via acquisition of specific genetic alterations that interfere with the shared mitochondrial death signaling pathway entrained by granzyme B, IFN-gamma, and Apo2 ligand/tumor necrosis factor-related apoptosis inducing ligand (Apo2L/TRAIL), three key mediators of immunologic cell-mediated cytotoxicity. We show that the coexistence of specific mitochondrial signaling defects (either deletion of Bax, overexpression of Bcl-x(L), or deletion of Smac) with expression of X-linked inhibitor of apoptosis protein decreases the sensitivity of cancer cells to IFN-gamma/Apo2L/TRAIL- or granzyme B-induced apoptosis, lymphocyte-mediated cytotoxicity in vitro, and adoptive cellular immunotherapy in vivo. Conversely, negating X-linked inhibitor of apoptosis protein expression or function in tumor cells with defective mitochondrial signaling enables direct activation of caspase-3/-7 by granzyme B or Apo2L/TRAIL, and restores their susceptibility to immunologic cytotoxicity. These findings identify an important mechanism by which cancers evade elimination by immune effector cells and suggest that cancer immunotherapy might be improved by concurrent strategies to alleviate or circumvent the intrinsic mitochondrial death signaling defects that help cancer cells resist immunologic cytotoxicity.
Collapse
Affiliation(s)
- Rajani Ravi
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
287
|
Holcík M. Targeting endogenous inhibitors of apoptosis for treatment of cancer, stroke and multiple sclerosis. Expert Opin Ther Targets 2006; 8:241-53. [PMID: 15161430 DOI: 10.1517/14728222.8.3.241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The inhibitor of apoptosis (IAP) genes have emerged as probably the most important intrinsic regulators of apoptosis. The members of the IAP family are highly conserved in evolutionarily distant species and perform the critical role of binding to and inhibiting distinct caspases. This inhibition is mediated by discrete baculoviral IAP repeat domains that, in a domain-specific manner, inhibit either the initiator or executioner caspases. As such the function of IAPs lies at the very centre of virtually all apoptotic pathways. Since many, if not most, human pathologies involve aberrant apoptosis, the modulation of IAP levels or their activity offers huge therapeutic potential for treatment of various disorders. Indeed, available data suggest that the therapeutic downregulation of IAPs by antisense targeting or their adenovirally-mediated overexpression, can in fact be used to successfully modulate cell death.
Collapse
Affiliation(s)
- Martin Holcík
- Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute and Department of Pediatrics, University of Ottawa, 401 Smyth Road, Ottawa, Ontario, K1H 8L1, USA.
| |
Collapse
|
288
|
Abstract
Apoptosis plays a central role in the development and homeostasis of metazoans. Research in the past two decades has led to the identification of hundreds of genes that govern the initiation, execution, and regulation of apoptosis. An earlier focus on the genetic and cell biological characterization has now been complemented by systematic biochemical and structural investigation, giving rise to an unprecedented level of clarity in many aspects of apoptosis. In this review, we focus on the molecular mechanisms of apoptosis by synthesizing available biochemical and structural information. We discuss the mechanisms of ligand binding to death receptors, actions of the Bcl-2 family of proteins, and caspase activation, inhibition, and removal of inhibition. Although an emphasis is given to the mammalian pathways, a comparative analysis is applied to related mechanistic information in Drosophila and Caenorhabditis elegans.
Collapse
Affiliation(s)
- Nieng Yan
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey 08544, USA.
| | | |
Collapse
|
289
|
Kamada S, Kikkawa U, Tsujimoto Y, Hunter T. A-kinase-anchoring protein 95 functions as a potential carrier for the nuclear translocation of active caspase 3 through an enzyme-substrate-like association. Mol Cell Biol 2005; 25:9469-77. [PMID: 16227597 PMCID: PMC1265837 DOI: 10.1128/mcb.25.21.9469-9477.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Caspase-mediated proteolysis is a critical and central element of the apoptotic process, and caspase 3, one of the effector caspases, is proposed to play essential roles in the nuclear morphological changes of apoptotic cells. Although many substrates for caspase 3 localize in the nucleus and caspase 3 translocates from the cytoplasm to the nuclei after activation in apoptotic cells, the molecular mechanisms of nuclear translocation of active caspase 3 have been unclear. Recently, we suggested that a substrate-like protein(s) served as a carrier to transport caspase 3 from the cytoplasm into the nucleus. In the present study, we identified A-kinase-anchoring protein 95 (AKAP95) as a caspase 3-binding protein. Small interfering RNA-mediated depletion of AKAP95 reduced apoptotic nuclear morphological changes, suggesting that AKAP95 is involved in the process of apoptotic nuclear morphological changes. The association of AKAP95 with active caspase 3 was analogous to an enzyme-substrate interaction. Furthermore, overexpression of AKAP95 with nuclear localization sequence mutations inhibited nuclear morphological changes in apoptotic cells. These results indicate that AKAP95 is a potential carrier protein for active caspase 3 from the cytoplasm into the nuclei in apoptotic cells.
Collapse
Affiliation(s)
- Shinji Kamada
- Molecular and Cell Biology Laboratory, The Salk Institute, La Jolla, CA, USA.
| | | | | | | |
Collapse
|
290
|
Guégan C, Braudeau J, Couriaud C, Dietz GPH, Lacombe P, Bähr M, Nosten-Bertrand M, Onténiente B. PTD-XIAP protects against cerebral ischemia by anti-apoptotic and transcriptional regulatory mechanisms. Neurobiol Dis 2005; 22:177-86. [PMID: 16361106 DOI: 10.1016/j.nbd.2005.10.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 10/20/2005] [Accepted: 10/27/2005] [Indexed: 12/31/2022] Open
Abstract
Caspases play a major role in the infarction process that follows occlusion of cerebral arteries and are important targets for stroke therapy. We have generated three fusion proteins that link various domains of the X chromosome-linked inhibitor of apoptosis (XIAP), a potent caspase inhibitor, to the protein transduction domain (PTD) of HIV-1/Tat, and have tested their efficacy after distal occlusion of the middle cerebral artery (dMCAO) in mice. PTD-XIAP failed to accumulate in brain structures after intravenous (iv) delivery, but properly transduced cortical cells when applied topically. Shorter constructs efficiently targeted the lesion after iv delivery. All proteins retained their caspase inhibitory activity and significantly reduced infarct volumes. PTD-XIAP reversed long-term impairments in the water maze test. Sequential activation of transcription factors was observed, suggesting that the effects of XIAP are mediated by both direct inhibition of apoptotic mechanisms and secondary regulation of transcription factors involved in neuronal survival.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Brain Ischemia/drug therapy
- Brain Ischemia/genetics
- Brain Ischemia/metabolism
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Cerebral Cortex/physiopathology
- Cerebral Infarction/drug therapy
- Cerebral Infarction/physiopathology
- Cerebral Infarction/prevention & control
- Disease Models, Animal
- Gene Products, tat/genetics
- Gene Products, tat/pharmacology
- Gene Products, tat/therapeutic use
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/metabolism
- Infusion Pumps
- Male
- Maze Learning/drug effects
- Maze Learning/physiology
- Mice
- Mice, Inbred C57BL
- Protein Structure, Tertiary/genetics
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/therapeutic use
- Regulatory Elements, Transcriptional/drug effects
- Regulatory Elements, Transcriptional/genetics
- Transcriptional Activation/drug effects
- Transcriptional Activation/physiology
- X-Linked Inhibitor of Apoptosis Protein/genetics
- X-Linked Inhibitor of Apoptosis Protein/pharmacology
- X-Linked Inhibitor of Apoptosis Protein/therapeutic use
Collapse
Affiliation(s)
- Christelle Guégan
- INSERM UMR421, Universite Paris 12, 8, rue du Général Sarrail, F-94010 Creteil cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
291
|
Lavrik IN, Golks A, Krammer PH. Caspases: pharmacological manipulation of cell death. J Clin Invest 2005; 115:2665-72. [PMID: 16200200 PMCID: PMC1236692 DOI: 10.1172/jci26252] [Citation(s) in RCA: 437] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Caspases, a family of cysteine proteases, play a central role in apoptosis. During the last decade, major progress has been made to further understand caspase structure and function, providing a unique basis for drug design. This Review gives an overview of caspases and their classification, structure, and substrate specificity. We also describe the current knowledge of how interference with caspase signaling can be used to pharmacologically manipulate cell death.
Collapse
Affiliation(s)
- Inna N Lavrik
- Division of Immunogenetics, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | | | | |
Collapse
|
292
|
Eckelman BP, Salvesen GS. The human anti-apoptotic proteins cIAP1 and cIAP2 bind but do not inhibit caspases. J Biol Chem 2005; 281:3254-60. [PMID: 16339151 DOI: 10.1074/jbc.m510863200] [Citation(s) in RCA: 289] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
cIAPs (cellular inhibitor of apoptosis proteins) 1 and 2 are able to regulate apoptosis when ectopically expressed in recipient cells and probably also in vivo. Previous work suggested that this is at least partially due to direct caspase inhibition, mediated by two of the three baculovirus IAP repeat (BIR) domains that are contained in these proteins. In support of this we show that the BIR domains 2 and 3 of the two cIAPs are able to bind caspases-7 and -9. However, we demonstrate that neither of these BIR domains is able to inhibit caspases because of critical substitutions in the regions that target caspase inhibition in the X-linked IAP, a tight binding caspase inhibitor. The cIAP BIR domains can be converted to tight binding caspase inhibitors by substituting these critical residues with XIAP residues. Thus, cIAPs maintain protein scaffolds suitable for direct caspase inhibition but have lost or never acquired specific caspase inhibitory interaction sites. Consequently, although the binding function of the cIAP BIRs may be important for their physiologic function, caspase inhibition is not.
Collapse
Affiliation(s)
- Brendan P Eckelman
- Program in Cell Death and Apoptosis Research, Burnham Institute for Medical Research and the Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, California 92037, USA
| | | |
Collapse
|
293
|
Bockbrader KM, Tan M, Sun Y. A small molecule Smac-mimic compound induces apoptosis and sensitizes TRAIL- and etoposide-induced apoptosis in breast cancer cells. Oncogene 2005; 24:7381-8. [PMID: 16044155 DOI: 10.1038/sj.onc.1208888] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inhibitor of apoptosis protein (IAP) suppresses apoptosis through binding and inhibiting active caspases-3, -7 and -9 via its baculoviral IAP repeat (BIR) domains. During apoptosis the caspase inhibition by IAPs can be negatively regulated by a mitochondrial protein second mitochondrial-derived activator of caspase (Smac). Smac physically interacts with multiple IAPs and relieves their inhibitory effect on caspases-3, -7 and -9. Recently, a small molecule Smac-mimic compound (Smac-mimic), which potentiates TNF-related apoptosis-inducing ligand (TRAIL) and tumor necrosis factor (TNF)-alpha mediated cell death in glioblastoma T98G cells and HeLa cells, was identified and characterized. To determine the efficacy of this compound in breast cancer cells, we first measured protein expression of three IAPs: XIAP, cIAP-1, and cIAP-2 in nine independent breast cancer cell lines. Three cell lines were chosen: a high IAPs expressing line MDA-MB-231, and two low IAPs expressing lines, T47D and MDA-MB-453. The cell lines were tested for their sensitivity to Smac-mimic alone or in combination with TRAIL or etoposide. Acting alone, Smac-mimic was quite potent with a cytotoxic IC50 of 3.8 nM in high IAPs expressing MDA-MB-231 cells, but was inactive at a much higher concentration in low IAPs expressing T47D and MDA-MB-453 cells. In fact, as low as 2.5 nM of Smac-mimic alone was sufficient to activate caspase-3 and induce apoptosis in MDA-MB-231 cells. In combinational treatments with TRAIL or etoposide, Smac-mimic significantly sensitized cells to growth suppression in MDA-MB-231 cells, but to a lesser extent in T47D and MDA-MB-453 cells. Furthermore, it significantly synergized MDA-MB-231, but not T47D cells to apoptosis induced by either TRAIL or etoposide. Thus, in these cell lines, Smac-mimic acts in an apparent IAPs dependent manner to induce apoptosis alone as well as sensitizes breast cancer cells to TRAIL or etoposide induced apoptosis via caspase-3 activation.
Collapse
Affiliation(s)
- Katrina M Bockbrader
- Department of Radiation Oncology, Division of Cancer Biology, University of Michigan Comprehensive Cancer Center, 4304 CCGC, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0936, USA
| | | | | |
Collapse
|
294
|
Abstract
Apoptosis is a highly regulated process of cell deletion and plays a fundamental role in the maintenance of tissue homeostasis in the adult organism. Numerous studies in recent years have revealed that apoptosis is a constitutive suicide programme expressed in most, if not all cells, and can be triggered by a variety of extrinsic and intrinsic signals. Many human diseases can be attributed directly or indirectly to a derangement of apoptosis, resulting in either cell accumulation, in which cell eradication or cell turnover is impaired, or cell loss, in which the apoptotic programme is inadvertently triggered. In addition, defective macrophage engulfment and degradation of cell corpses may also contribute to a dysregulation of tissue homeostasis. An increased understanding of the signalling pathways that govern the execution of apoptosis and the subsequent clearance of dying cells may thus yield novel targets for therapeutic intervention in a wide range of human maladies.
Collapse
Affiliation(s)
- B Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
295
|
Zhang N, Hartig H, Dzhagalov I, Draper D, He YW. The role of apoptosis in the development and function of T lymphocytes. Cell Res 2005; 15:749-69. [PMID: 16246265 DOI: 10.1038/sj.cr.7290345] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Apoptosis plays an essential role in T cell biology. Thymocytes expressing nonfunctional or autoreactive TCRs are eliminated by apoptosis during development. Apoptosis also leads to the deletion of expanded effector T cells during immune responses. The dysregulation of apoptosis in the immune system results in autoimmunity, tumorogenesis and immunodeficiency. Two major pathways lead to apoptosis: the intrinsic cell death pathway controlled by Bcl-2 family members and the extrinsic cell death pathway controlled by death receptor signaling. These two pathways work together to regulate T lymphocyte development and function.
Collapse
Affiliation(s)
- Nu Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
296
|
Cashio P, Lee TV, Bergmann A. Genetic control of programmed cell death in Drosophila melanogaster. Semin Cell Dev Biol 2005; 16:225-35. [PMID: 15797833 DOI: 10.1016/j.semcdb.2005.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Apoptosis is a genetically controlled form of cell death that is an important feature of animal development and homeostasis. The genes involved in the control and execution of apoptosis are conserved throughout evolution. However, the actual molecular mechanisms used by these genes vary from species to species. In this review, we focus on the genetic components of apoptosis in the fruit fly Drosophila melanogaster, and compare their mode of action to the one employed by the homologous genes in mammals. We also cover recent advances that show that apoptotic genes have a requirement in processes other than apoptosis.
Collapse
Affiliation(s)
- Peter Cashio
- The University of Texas, MD Anderson Cancer Center, Department of Biochemistry and Molecular Biology, The Genes and Development Graduate Program, Houston, TX 77030, USA
| | | | | |
Collapse
|
297
|
Siskind LJ, Fluss S, Bui M, Colombini M. Sphingosine forms channels in membranes that differ greatly from those formed by ceramide. J Bioenerg Biomembr 2005; 37:227-36. [PMID: 16167178 PMCID: PMC2222862 DOI: 10.1007/s10863-005-6632-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 03/22/2005] [Indexed: 10/25/2022]
Abstract
Ceramide channels formed in the outer membrane of mitochondria have been proposed to be the pathways by which proapoptotic proteins are released from mitochondria during the early stages of apoptosis. We report that sphingosine also forms channels in membranes, but these differ greatly from the large oligomeric barrel-stave channels formed by ceramide. Sphingosine channels have short open lifetimes and have diameters less than 2 nm, whereas ceramide channels have long open lifetimes, enlarge in size reaching diameters in excess of 10 nm. Unlike ceramide, sphingosine forms channels in erythrocyte plasma membranes that vary in size with concentration, but with a maximum possible channel diameter of 2 nm. In isolated mitochondria, a large proportion of the added sphingosine was rapidly metabolized to ceramide in the absence of externally added fatty acids or fatty-acyl-CoAs. The ceramide synthase inhibitor, fumonisin B1 failed to prevent sphingosine metabolism to ceramide and actually increased it. However, partial inhibition of conversion to ceramide was achieved in the presence of ceramidase inhibitors, indicating that reverse ceramidase activity is at least partially responsible for sphingosine metabolism to ceramide. A small amount of cytochrome c release was detected. It correlated with the level of ceramide converted from sphingosine. Thus, sphingosine channels, unlike ceramide channels, are not large enough to allow the passage of proapoptotic proteins from the intermembrane space of mitochondria to the cytoplasm.
Collapse
Affiliation(s)
- Leah J. Siskind
- Department of Biology, University of Maryland College Park, College Park, Maryland
| | - Sharon Fluss
- Department of Biology, University of Maryland College Park, College Park, Maryland
| | - Minh Bui
- Department of Biology, University of Maryland College Park, College Park, Maryland
| | - Marco Colombini
- Department of Biology, University of Maryland College Park, College Park, Maryland
| |
Collapse
|
298
|
Shin H, Renatus M, Eckelman B, Nunes V, Sampaio C, Salvesen G. The BIR domain of IAP-like protein 2 is conformationally unstable: implications for caspase inhibition. Biochem J 2005; 385:1-10. [PMID: 15485395 PMCID: PMC1134667 DOI: 10.1042/bj20041107] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Several IAP (inhibitor of apoptosis) proteins regulate cell fate decisions, and the X-linked IAP (XIAP) does so in part by inhibiting caspases, proteases that execute the apoptotic pathway. A tissue-specific homologue of XIAP, known as ILP2 (IAP-like protein 2), has previously been implicated in the control of apoptosis in the testis by direct inhibition of caspase 9. In examining this protein we found that the putative caspase 9 interaction domain is a surprisingly weak inhibitor and is also conformationally unstable. Comparison with the equivalent domain in XIAP demonstrated that the instability is due to the lack of a linker segment N-terminal to the inhibitory BIR (baculovirus IAP repeat) domain. Fusion of a 9-residue linker from XIAP to the N-terminus of ILP2 restored tight caspase 9 inhibition, dramatically increased conformational stability and allowed crystallization of the ILP2 BIR domain in a form strikingly similar to the XIAP third BIR domain. We conclude that ILP2 is an unstable protein, and cannot inhibit caspase 9 in a physiological way on its own. We speculate that ILP2 requires assistance from unidentified cellular factors to be an effective inhibitor of apoptosis in vivo.
Collapse
Affiliation(s)
- Hwain Shin
- *Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
- †Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, CA 92037, U.S.A
| | - Martin Renatus
- *Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Brendan P. Eckelman
- *Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
- †Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, CA 92037, U.S.A
| | - Viviane A. Nunes
- *Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
- ‡Department of Biochemistry, Escola Paulista de Medicina, UNIFESP, Sao Paulo, SP, Brazil
| | - Claudio A. M. Sampaio
- ‡Department of Biochemistry, Escola Paulista de Medicina, UNIFESP, Sao Paulo, SP, Brazil
| | - Guy S. Salvesen
- *Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
299
|
Vucic D, Franklin M, Wallweber H, Das K, Eckelman B, Shin H, Elliott L, Kadkhodayan S, Deshayes K, Salvesen G, Fairbrother W. Engineering ML-IAP to produce an extraordinarily potent caspase 9 inhibitor: implications for Smac-dependent anti-apoptotic activity of ML-IAP. Biochem J 2005; 385:11-20. [PMID: 15485396 PMCID: PMC1134668 DOI: 10.1042/bj20041108] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ML-IAP (melanoma inhibitor of apoptosis) is a potent anti-apoptotic protein that is strongly up-regulated in melanoma and confers protection against a variety of pro-apoptotic stimuli. The mechanism by which ML-IAP regulates apoptosis is unclear, although weak inhibition of caspases 3 and 9 has been reported. Here, the binding to and inhibition of caspase 9 by the single BIR (baculovirus IAP repeat) domain of ML-IAP has been investigated and found to be significantly less potent than the ubiquitously expressed XIAP (X-linked IAP). Engineering of the ML-IAP-BIR domain, based on comparisons with the third BIR domain of XIAP, resulted in a chimeric BIR domain that binds to and inhibits caspase 9 significantly better than either ML-IAP-BIR or XIAP-BIR3. Mutational analysis of the ML-IAP-BIR domain demonstrated that similar enhancements in caspase 9 affinity can be achieved with only three amino acid substitutions. However, none of these modifications affected binding of the ML-IAP-BIR domain to the IAP antagonist Smac (second mitochondrial activator of caspases). ML-IAP-BIR was found to bind mature Smac with low nanomolar affinity, similar to that of XIAP-BIR2-BIR3. Correspondingly, increased expression of ML-IAP results in formation of a ML-IAP-Smac complex and disruption of the endogenous interaction between XIAP and mature Smac. These results suggest that ML-IAP might regulate apoptosis by sequestering Smac and preventing it from antagonizing XIAP-mediated inhibition of caspases, rather than by direct inhibition of caspases.
Collapse
Affiliation(s)
- Domagoj Vucic
- *Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Matthew C. Franklin
- †Department of Protein Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Heidi J. A. Wallweber
- †Department of Protein Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Kanad Das
- †Department of Protein Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Brendan P. Eckelman
- ‡Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, CA 92037, U.S.A
- §Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Hwain Shin
- ‡Graduate Program in Molecular Pathology, University of California San Diego, La Jolla, CA 92037, U.S.A
| | - Linda O. Elliott
- ∥Department of Medicinal Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Saloumeh Kadkhodayan
- ¶Department of Bioanalytical Research and Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Kurt Deshayes
- †Department of Protein Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
| | - Guy S. Salvesen
- §Program in Apoptosis and Cell Death Research, The Burnham Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Wayne J. Fairbrother
- *Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
- †Department of Protein Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
300
|
Abstract
In most cell types, a key event in apoptosis is the release of proapoptotic intermembrane space proteins from mitochondria to the cytoplasm. In general, it is the release of these intermembrane space proteins that is responsible for the activation of caspases and DNases that are responsible for the execution of apoptosis. The mechanism for the increased permeability of the mitochondrial outer membrane during the induction phase of apoptosis is currently unknown and highly debated. This review will focus on one such proposed mechanism, namely, the formation of ceramide channels in the mitochondrial outer membrane. Ceramides are known to play a major regulatory role in apoptosis by inducing the release of proapoptotic proteins from the mitochondria. As mitochondria are known to contain the enzymes responsible for the synthesis and hydrolysis of ceramide, there exists a mechanism for regulating the level of ceramide in mitochondria. In addition, mitochondrial ceramide levels have been shown to be elevated prior to the induction phase of apoptosis. Ceramide has been shown to form large protein permeable channels in planar phospholipid and mitochondrial outer membranes. Thus, ceramide channels are good candidates for the pathway with which proapoptotic proteins are released from mitochondria during the induction phase of apoptosis.
Collapse
Affiliation(s)
- Leah J Siskind
- Department of Biology, University of Maryland-College Park, College Park, MD 20742, USA.
| |
Collapse
|