201
|
Apoptosis: the intrinsic pathway. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
202
|
Morishita J, Kang MJ, Fidelin K, Ryoo HD. CDK7 regulates the mitochondrial localization of a tail-anchored proapoptotic protein, Hid. Cell Rep 2013; 5:1481-8. [PMID: 24360962 DOI: 10.1016/j.celrep.2013.11.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/26/2013] [Accepted: 11/15/2013] [Indexed: 01/20/2023] Open
Abstract
The mitochondrial outer membrane is a major site of apoptosis regulation across phyla. Human and C. elegans Bcl-2 family proteins and Drosophila Hid require the C-terminal tail-anchored (TA) sequence in order to insert into the mitochondrial membrane, but it remains unclear whether cytosolic proteins actively regulate the mitochondrial localization of these proteins. Here, we report that the cdk7 complex regulates the mitochondrial localization of Hid and its ability to induce apoptosis. We identified cdk7 through an in vivo RNAi screen of genes required for cell death. Although CDK7 is best known for its role in transcription and cell-cycle progression, a hypomorphic cdk7 mutant suppressed apoptosis without impairing these other known functions. In this cdk7 mutant background, Hid failed to localize to the mitochondria and failed to bind to recombinant inhibitors of apoptosis (IAPs). These findings indicate that apoptosis is promoted by a newly identified function of CDK7, which couples the mitochondrial localization and IAP binding of Hid.
Collapse
Affiliation(s)
- Jun Morishita
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Min-Ji Kang
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Kevin Fidelin
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
203
|
Hennessy EJ, Adam A, Aquila BM, Castriotta LM, Cook D, Hattersley M, Hird AW, Huntington C, Kamhi VM, Laing NM, Li D, MacIntyre T, Omer CA, Oza V, Patterson T, Repik G, Rooney MT, Saeh JC, Sha L, Vasbinder MM, Wang H, Whitston D. Discovery of a Novel Class of Dimeric Smac Mimetics as Potent IAP Antagonists Resulting in a Clinical Candidate for the Treatment of Cancer (AZD5582). J Med Chem 2013; 56:9897-919. [DOI: 10.1021/jm401075x] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Edward J. Hennessy
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ammar Adam
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Brian M. Aquila
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Lillian M. Castriotta
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Donald Cook
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Maureen Hattersley
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Alexander W. Hird
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Christopher Huntington
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Victor M. Kamhi
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Naomi M. Laing
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Danyang Li
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Terry MacIntyre
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Charles A. Omer
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Vibha Oza
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Troy Patterson
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Galina Repik
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Michael T. Rooney
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jamal C. Saeh
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Li Sha
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Melissa M. Vasbinder
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Haiyun Wang
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - David Whitston
- Oncology iMed, Innovative Medicines & Early Development, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
204
|
Suwannaboon R, Phiwsaiya K, Senapin S, Khunrae P, Rattanarojpong T. The identification and expression of the full-length HtrA2 gene from Penaeus monodon (black tiger shrimp). Protein Expr Purif 2013; 92:183-9. [DOI: 10.1016/j.pep.2013.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/14/2013] [Accepted: 09/18/2013] [Indexed: 11/29/2022]
|
205
|
Shiokawa Z, Hashimoto K, Saito B, Oguro Y, Sumi H, Yabuki M, Yoshimatsu M, Kosugi Y, Debori Y, Morishita N, Dougan DR, Snell GP, Yoshida S, Ishikawa T. Design, synthesis, and biological activities of novel hexahydropyrazino[1,2-a]indole derivatives as potent inhibitors of apoptosis (IAP) proteins antagonists with improved membrane permeability across MDR1 expressing cells. Bioorg Med Chem 2013; 21:7938-54. [DOI: 10.1016/j.bmc.2013.09.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/25/2013] [Accepted: 09/27/2013] [Indexed: 02/02/2023]
|
206
|
Li J, Wang Y, DU L, Xu C, Cao J, Wang Q, Liu Q, Fan F. Radiation-induced cytochrome c release and the neuroprotective effects of the pan-caspase inhibitor z-VAD-fmk in the hypoglossal nucleus. Exp Ther Med 2013; 7:383-388. [PMID: 24396410 PMCID: PMC3881038 DOI: 10.3892/etm.2013.1419] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 11/04/2013] [Indexed: 02/02/2023] Open
Abstract
Numerous studies have demonstrated that neuronal cell death occurs via extrinsic (death receptors) and intrinsic (mitochondria) pathways. Radiation induces caspase activation fundamentally via the mitochondrial pathway. To investigate the role of caspase, a cell permeable pan-caspase inhibitor, z-VAD-fmk [N-benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone], was used to investigate the effects of caspase blockade in vivo following irradiation. Adult male Sprague-Dawley rats (weight, 250–300 g) underwent irradiation at room temperature with a 4-Gy dose of radiation. Since z-VAD-fmk does not penetrate the blood-brain barrier, it was applied intracerebroventricularly via a bolus injection (0.2 μg/h for 1 h). Terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) demonstrated that z-VAD-fmk reduced the numbers of TUNEL-positive cells within the hypoglossal nucleus, suggesting that intervention in the caspase cascade following radiation may have therapeutic applications. The caspase inhibitor z-VAD-fmk reduced the expression and activation of caspase-3, caspase-8 and caspase-9 in the irradiated rats, indicating that caspase may be a potential therapeutic target in the treatment of brain radiation injury. Treatment with z-VAD-fmk also reduced the appearance of cytochrome c within the cytosolic fraction following radiation. The hypoglossal nucleus may be used as a model of radiation-induced injury in the central nervous system, providing visual information and displaying apoptotic nuclear morphology.
Collapse
Affiliation(s)
- Jianguo Li
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China ; Department of Human Anatomy, The Medical School of Inner Mongolia University for the Nationalities, Tongliao, Neimenggu 028041, P.R. China
| | - Yan Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Liqing DU
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Chang Xu
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Jia Cao
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Qin Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Qiang Liu
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| | - Feiyue Fan
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Nankai, Tianjin 300192, P.R. China
| |
Collapse
|
207
|
Kester RF, Donnell AF, Lou Y, Remiszewski SW, Lombardo LJ, Chen S, Le NT, Lo J, Moliterni JA, Han X, Hogg JH, Liang W, Michoud C, Rupert KC, Mischke S, Le K, Weisel M, Janson CA, Lukacs CM, Fretland AJ, Hong K, Polonskaia A, Gao L, Li S, Solis DS, Aguilar D, Tardell C, Dvorozniak M, Tannu S, Lee EC, Schutt AD, Goggin B. Optimization of benzodiazepinones as selective inhibitors of the X-linked inhibitor of apoptosis protein (XIAP) second baculovirus IAP repeat (BIR2) domain. J Med Chem 2013; 56:7788-803. [PMID: 24093940 DOI: 10.1021/jm400732v] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The IAPs are key regulators of the apoptotic pathways and are commonly overexpressed in many cancer cells. IAPs contain one to three BIR domains that are crucial for their inhibitory function. The pro-survival properties of XIAP come from binding of the BIR domains to the pro-apoptotic caspases. The BIR3 domain of XIAP binds and inhibits caspase 9, while the BIR2 domain binds and inhibits the terminal caspases 3 and 7. While XIAP BIR3 inhibitors have previously been reported, they also inhibit cIAP1/2 and promote the release of TNFα, potentially limiting their therapeutic utility. This paper will focus on the optimization of selective XIAP BIR2 inhibitors leading to the discovery of highly potent benzodiazepinone 36 (IC50 = 45 nM), which has high levels of selectivity over XIAP BIR3 and cIAP1 BIR2/3 and shows efficacy in a xenograft pharmacodynamic model monitoring caspase activity while not promoting the release of TNFα in vitro.
Collapse
Affiliation(s)
- Robert F Kester
- Departments of Discovery Chemistry, ‡Discovery Technologies, §Non-clinical Safety, Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc. , 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Xue X, Wei JL, Xu LL, Xi MY, Xu XL, Liu F, Guo XK, Wang L, Zhang XJ, Zhang MY, Lu MC, Sun HP, You QD. Effective screening strategy using ensembled pharmacophore models combined with cascade docking: application to p53-MDM2 interaction inhibitors. J Chem Inf Model 2013; 53:2715-29. [PMID: 24050442 DOI: 10.1021/ci400348f] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Protein-protein interactions (PPIs) play a crucial role in cellular function and form the backbone of almost all biochemical processes. In recent years, protein-protein interaction inhibitors (PPIIs) have represented a treasure trove of potential new drug targets. Unfortunately, there are few successful drugs of PPIIs on the market. Structure-based pharmacophore (SBP) combined with docking has been demonstrated as a useful Virtual Screening (VS) strategy in drug development projects. However, the combination of target complexity and poor binding affinity prediction has thwarted the application of this strategy in the discovery of PPIIs. Here we report an effective VS strategy on p53-MDM2 PPI. First, we built a SBP model based on p53-MDM2 complex cocrystal structures. The model was then simplified by using a Receptor-Ligand complex-based pharmacophore model considering the critical binding features between MDM2 and its small molecular inhibitors. Cascade docking was subsequently applied to improve the hit rate. Based on this strategy, we performed VS on NCI and SPECS databases and successfully discovered 6 novel compounds from 15 hits with the best, compound 1 (NSC 5359), K(i) = 180 ± 50 nM. These compounds can serve as lead compounds for further optimization.
Collapse
Affiliation(s)
- Xin Xue
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing, Jiangsu 210009, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Donnell AF, Michoud C, Rupert KC, Han X, Aguilar D, Frank KB, Fretland AJ, Gao L, Goggin B, Hogg JH, Hong K, Janson CA, Kester RF, Kong N, Le K, Li S, Liang W, Lombardo LJ, Lou Y, Lukacs CM, Mischke S, Moliterni JA, Polonskaia A, Schutt AD, Solis DS, Specian A, Taylor RT, Weisel M, Remiszewski SW. Benzazepinones and Benzoxazepinones as Antagonists of Inhibitor of Apoptosis Proteins (IAPs) Selective for the Second Baculovirus IAP Repeat (BIR2) Domain. J Med Chem 2013; 56:7772-87. [DOI: 10.1021/jm400731m] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Andrew F. Donnell
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Christophe Michoud
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Kenneth C. Rupert
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Xiaochun Han
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Douglas Aguilar
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Karl B. Frank
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Adrian J. Fretland
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Lin Gao
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Barry Goggin
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - J. Heather Hogg
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Kyoungja Hong
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Cheryl A. Janson
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Robert F. Kester
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Norman Kong
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Kang Le
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Shirley Li
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Weiling Liang
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Louis J. Lombardo
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Yan Lou
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Christine M. Lukacs
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Steven Mischke
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - John A. Moliterni
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Ann Polonskaia
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Andrew D. Schutt
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Dave S. Solis
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Anthony Specian
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Robert T. Taylor
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Martin Weisel
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| | - Stacy W. Remiszewski
- Departments of Discovery
Chemistry, ‡Discovery Technologies, §Non-clinical Safety,
Early ADME, and ∥Discovery Oncology, Hoffmann-La Roche Inc., 340 Kingsland Street, Nutley, New Jersey 07110, United States
| |
Collapse
|
210
|
Sleiman L, Beanlands R, Hasu M, Thabet M, Norgaard A, Chen YX, Holcik M, Whitman S. Loss of cellular inhibitor of apoptosis protein 2 reduces atherosclerosis in atherogenic apoE-/- C57BL/6 mice on high-fat diet. J Am Heart Assoc 2013; 2:e000259. [PMID: 24072531 PMCID: PMC3835229 DOI: 10.1161/jaha.113.000259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Cellular inhibitor of apoptosis protein 2 (cIAP2) is predicted to participate in atherosclerosis; however, its direct role in atherosclerosis development has not been investigated. We aimed to examine and assess the loss of cIAP2 on atherosclerosis lesion development. Methods and Results We used apoE−/− C57BL/6 male mice, either cIAP2−/− or cIAP2+/+. At 8 weeks, mice were fed a high‐fat diet (HFD) for 4 and 12 weeks. Aortic root was serially sectioned and stained with Sudan IV, CD68, α‐actin, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). cIAP2−/− mice displayed a significant decrease in atherosclerotic lesion's macrophage number after 4 weeks of HFD. Similarly, decrease in lesion area at 4 and 12 weeks HFD was detected by use of en face analysis (cIAP2−/− 0.58±0.37% versus cIAP2+/+ 1.51±0.79% [P=0.0056]); (cIAP2−/− 9.34±4.88% versus cIAP2+/+ 17.65±6.24% [P=0.0019]). Aortic root lesion area after 4 and 12 weeks of HFD also decreased (cIAP2−/− 0.0328±0.014 mm2 versus cIAP2+/+ 0.0515±0.021 mm2 [P=0.022]); (cIAP2−/− 0.3614±0.1157 mm2 versus cIAP2+/+ 0.4901±0.125 mm2 [P=0.065]). TUNEL analysis after 4 and 12 weeks of HFD showed a 2.5‐fold increase in TUNEL+ cells (cIAP2−/− 4.47±2.26% versus cIAP2+/+ 1.74±0.98% [P=0.036]); (cIAP2−/− 2.39±0.75% versus cIAP2+/+ 1.29±0.47% [P=0.032]). Smooth muscle cell content in cIAP2−/− mice was 3.075±3.3% compared with cIAP2+/+ with 0.085±0.1% (P=0.0071). Conclusions Results uncover a key role for cIAP2 in atherosclerotic lesion development, and targeting it may represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Lyne Sleiman
- Departments of Pathology and Laboratory Medicine and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Dubrez L, Berthelet J, Glorian V. IAP proteins as targets for drug development in oncology. Onco Targets Ther 2013; 9:1285-304. [PMID: 24092992 PMCID: PMC3787928 DOI: 10.2147/ott.s33375] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The inhibitors of apoptosis (IAPs) constitute a family of proteins involved in the regulation of
various cellular processes, including cell death, immune and inflammatory responses, cell
proliferation, cell differentiation, and cell motility. There is accumulating evidence supporting
IAP-targeting in tumors: IAPs regulate various cellular processes that contribute to tumor
development, such as cell death, cell proliferation, and cell migration; their expression is
increased in a number of human tumor samples, and IAP overexpression has been correlated with tumor
growth, and poor prognosis or low response to treatment; and IAP expression can be rapidly induced
in response to chemotherapy or radiotherapy because of the presence of an internal ribosome entry
site (IRES)-dependent mechanism of translation initiation, which could contribute to resistance to
antitumor therapy. The development of IAP antagonists is an important challenge and was subject to
intense research over the past decade. Six molecules are currently in clinical trials. This review
focuses on the role of IAPs in tumors and the development of IAP-targeting molecules for anticancer
therapy.
Collapse
Affiliation(s)
- Laurence Dubrez
- Institut National de la Santé et de la Recherche Médicale (Inserm), Dijon, France ; Université de Bourgogne, Dijon, France
| | | | | |
Collapse
|
212
|
IAPs on the move: role of inhibitors of apoptosis proteins in cell migration. Cell Death Dis 2013; 4:e784. [PMID: 24008728 PMCID: PMC3789170 DOI: 10.1038/cddis.2013.311] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/09/2013] [Indexed: 01/06/2023]
Abstract
Inhibitors of Apoptosis Proteins (IAPs) are a class of highly conserved proteins predominantly known for the regulation of caspases and immune signaling. However, recent evidence suggests a crucial role for these molecules in the regulation of tumor cell shape and migration by controlling MAPK, NF-κB and Rho GTPases. IAPs directly control Rho GTPases, thus regulating cell shape and migration. For instance, XIAP and cIAP1 function as the direct E3 ubiquitin ligases of Rac1 and target it for proteasomal degradation. IAPs are differentially expressed in tumor cells and have been targeted by several cancer therapeutic drugs that are currently in clinical trials. Here, we summarize the current knowledge on the role of IAPs in the regulation of cell migration and discuss the possible implications of these observations in regulating tumor cell metastases.
Collapse
|
213
|
Jeong CH, Chun KS, Kundu J, Park B. Phosphorylation of Smac by Akt promotes the caspase-3 activation during etoposide-induced apoptosis in HeLa cells. Mol Carcinog 2013; 54:83-92. [DOI: 10.1002/mc.22075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Chul-Ho Jeong
- College of Pharmacy; Keimyung University; Daegu Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy; Keimyung University; Daegu Republic of Korea
| | - Juthika Kundu
- College of Pharmacy; Keimyung University; Daegu Republic of Korea
| | - Byoungduck Park
- College of Pharmacy; Keimyung University; Daegu Republic of Korea
| |
Collapse
|
214
|
Elsawy MA, Martin L, Tikhonova IG, Walker B. Solid phase synthesis of Smac/DIABLO-derived peptides using a ‘Safety-Catch’ resin: Identification of potent XIAP BIR3 antagonists. Bioorg Med Chem 2013; 21:5004-11. [DOI: 10.1016/j.bmc.2013.06.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 10/26/2022]
|
215
|
Lukacs C, Belunis C, Crowther R, Danho W, Gao L, Goggin B, Janson CA, Li S, Remiszewski S, Schutt A, Thakur MK, Singh SK, Swaminathan S, Pandey R, Tyagi R, Gosu R, Kamath AV, Kuglstatter A. The structure of XIAP BIR2: understanding the selectivity of the BIR domains. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:1717-25. [PMID: 23999295 PMCID: PMC3760131 DOI: 10.1107/s0907444913016284] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/11/2013] [Indexed: 11/11/2022]
Abstract
XIAP, a member of the inhibitor of apoptosis family of proteins, is a critical regulator of apoptosis. Inhibition of the BIR domain-caspase interaction is a promising approach towards treating cancer. Previous work has been directed towards inhibiting the BIR3-caspase-9 interaction, which blocks the intrinsic apoptotic pathway; selectively inhibiting the BIR2-caspase-3 interaction would also block the extrinsic pathway. The BIR2 domain of XIAP has successfully been crystallized; peptides and small-molecule inhibitors can be soaked into these crystals, which diffract to high resolution. Here, the BIR2 apo crystal structure and the structures of five BIR2-tetrapeptide complexes are described. The structural flexibility observed on comparing these structures, along with a comparison with XIAP BIR3, affords an understanding of the structural elements that drive selectivity between BIR2 and BIR3 and which can be used to design BIR2-selective inhibitors.
Collapse
Affiliation(s)
- Christine Lukacs
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Charles Belunis
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Robert Crowther
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Waleed Danho
- Medicinal Chemistry, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Lin Gao
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Barry Goggin
- Discovery Oncology, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Cheryl A. Janson
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Shirley Li
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Stacy Remiszewski
- Medicinal Chemistry, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | - Andrew Schutt
- Discovery Oncology, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| | | | - Saroj K. Singh
- Structural Biology, Jubilant Biosys Ltd, Bangalore, India
| | | | - Rajat Pandey
- Structural Biology, Jubilant Biosys Ltd, Bangalore, India
| | - Rajiv Tyagi
- Structural Biology, Jubilant Biosys Ltd, Bangalore, India
| | | | | | - Andreas Kuglstatter
- Discovery Technologies, Hoffmann-La Roche, 340 Kingsland Street, Nutley, NJ 07110, USA
| |
Collapse
|
216
|
Wadegaonkar VP, Wadegaonkar PA. Withanone as an inhibitor of survivin: a potential drug candidate for cancer therapy. J Biotechnol 2013; 168:229-33. [PMID: 23994265 DOI: 10.1016/j.jbiotec.2013.08.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Abstract
Survivin, the smallest inhibitor of apoptosis protein, which has been reported to be highly expressed in almost all known cancers, plays a dual role in survival as well as the proliferation of cancer cells. It inhibits apoptosis by inhibiting caspases as well as facilitating mitosis by becoming a part of chromosomal passenger complex through its BIR5 domain. Docking studies carried out with herbal ligand withanone derived from roots of Withania somnifera have shown strong binding affinity of -19.1088 kJ/mol with BIR5 domain of survivin and in turn interferes with inhibitory action against caspases and may lead to apoptosis. Binding of withanone at BIR5 domain of survivin may also interfere with chromosomal passenger complex and lead to halt the mitotic process within the cancer cell. Docking studies support various experimental outcomes and suggest withanone as a potential candidate molecule in cancer therapy.
Collapse
Affiliation(s)
- Varsha P Wadegaonkar
- Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, India
| | | |
Collapse
|
217
|
Liu BH, Chen L, Li SR, Wang ZX, Cheng WG. Smac/DIABLO regulates the apoptosis of hypertrophic scar fibroblasts. Int J Mol Med 2013; 32:615-22. [PMID: 23857156 DOI: 10.3892/ijmm.2013.1442] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 06/10/2013] [Indexed: 11/06/2022] Open
Abstract
In abnormal skin wound healing, hypertrophic scars (HS) are characterized by excessive fibroblast hypercellularity and an overproduction of collagen, leading to atypical extracellular matrix (ECM) remodeling. Although the exact mechanisms of HS remain unclear, decreased HS fibroblast (HSFB) apoptosis and increased proliferation are evident in the development of HS. In this study, the contribution of the second mitochondria-derived activator of caspases/direct inhibitor of apoptosis protein (IAP)-binding protein with a low isoelectric point (pI) (Smac/DIABLO), an apoptosis-promoting protein released from the mitochondria, was investigated in human normal skin and HSFB cultures. The expression of Smac/DIABLO is usually decreased in many malignant tumors compared with normal tissues. Immunohistochemical analysis of skin tissues and the western blot analyses of fibroblasts revealed that the expression of Smac/DIABLO was lower in HS tissues compared with normal skin tissues. Of note, adenovirus-mediated Smac/DIABLO overexpression in the cultured HSFBs significantly reduced cell proliferation, as detected by the cell counting kit-8, and increased caspase-3 and -9 activity, as detected by spectrofluorimetry. In addition, it increased apoptosis, as detected by fluorescence-activated cell sorting (FACS). Furthermore, we found that the silencing of Smac with siRNA in the HSFBs induced a noticeable decrease in caspase-3 and -9 activity, leading to a significant reduction in apoptosis. In addition, the mRNA expression of type I and III pro-collagen detected in the HSFBs was significantly increased following the silencing of Smac with siRNA and was inhibited following Smac/DIABLO overexpression, as shown by real-time RT-PCR. In conclusion, Smac/DIABLO decreases the proliferation and increases the apoptosis of HSFBs. To our knowledge, the data from our study suggest for the first time that Smac/DIABLO is a novel therapeutic target for HS.
Collapse
Affiliation(s)
- Bao-Heng Liu
- Department of Plastic and Reconstructive Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | | | | | | | | |
Collapse
|
218
|
Damgaard RB, Fiil BK, Speckmann C, Yabal M, zur Stadt U, Bekker-Jensen S, Jost PJ, Ehl S, Mailand N, Gyrd-Hansen M. Disease-causing mutations in the XIAP BIR2 domain impair NOD2-dependent immune signalling. EMBO Mol Med 2013; 5:1278-95. [PMID: 23818254 PMCID: PMC3944466 DOI: 10.1002/emmm.201303090] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 06/01/2013] [Accepted: 06/03/2013] [Indexed: 01/22/2023] Open
Abstract
X-linked Inhibitor of Apoptosis (XIAP) is an essential ubiquitin ligase for pro-inflammatory signalling downstream of the nucleotide-binding oligomerization domain containing (NOD)-1 and -2 pattern recognition receptors. Mutations in XIAP cause X-linked lymphoproliferative syndrome type-2 (XLP2), an immunodeficiency associated with a potentially fatal deregulation of the immune system, whose aetiology is not well understood. Here, we identify the XIAP baculovirus IAP repeat (BIR)2 domain as a hotspot for missense mutations in XLP2. We demonstrate that XLP2-BIR2 mutations severely impair NOD1/2-dependent immune signalling in primary cells from XLP2 patients and in reconstituted XIAP-deficient cell lines. XLP2-BIR2 mutations abolish the XIAP-RIPK2 interaction resulting in impaired ubiquitylation of RIPK2 and recruitment of linear ubiquitin chain assembly complex (LUBAC) to the NOD2-complex. We show that the RIPK2 binding site in XIAP overlaps with the BIR2 IBM-binding pocket and find that a bivalent Smac mimetic compound (SMC) potently antagonises XIAP function downstream of NOD2 to limit signalling. These findings suggest that impaired immune signalling in response to NOD1/2 stimulation is a general defect in XLP2 and demonstrate that the XIAP BIR2-RIPK2 interaction may be targeted pharmacologically to modulate inflammatory signalling. The X-linked lymphoproliferative syndrome type-2 is an immunodeficiency disease caused by mutations in the XIAP gene. BIR2 domain mutations in patients impair RIPK2 binding and NOD2-dependent innate immune signaling, explaining some of the pathology.
Collapse
Affiliation(s)
- Rune Busk Damgaard
- Department of Disease Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Sheng R, Sun H, Liu L, Lu J, McEachern D, Wang G, Wen J, Min P, Du Z, Lu H, Kang S, Guo M, Yang D, Wang S. A potent bivalent Smac mimetic (SM-1200) achieving rapid, complete, and durable tumor regression in mice. J Med Chem 2013; 56:3969-79. [PMID: 23651223 DOI: 10.1021/jm400216d] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have designed, synthesized, and evaluated a series of new compounds based upon our previously reported bivalent Smac mimetics. This led to the identification of compound 12 (SM-1200), which binds to XIAP, cIAP1, and cIAP2 with Ki values of 0.5, 3.7, and 5.4 nM, respectively, inhibits cell growth in the MDA-MB-231 breast cancer and SK-OV-3 ovarian cancer cell lines with IC50 values of 11.0 and 28.2 nM, respectively. Compound 12 has a much improved pharmacokinetic profile over our previously reported bivalent Smac mimetics and is highly effective in induction of rapid and durable tumor regression in the MDA-MB-231 xenograft model. These data indicate that compound 12 is a promising Smac mimetic and warrants extensive evaluation as a potential candidate for clinical development.
Collapse
Affiliation(s)
- Rong Sheng
- Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Chaganti LK, Kuppili RR, Bose K. Intricate structural coordination and domain plasticity regulate activity of serine protease HtrA2. FASEB J 2013; 27:3054-66. [DOI: 10.1096/fj.13-227256] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Lalith K. Chaganti
- Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Raja Reddy Kuppili
- Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| | - Kakoli Bose
- Advanced Centre for Treatment, Research, and Education in Cancer (ACTREC)Tata Memorial CentreNavi MumbaiIndia
| |
Collapse
|
221
|
Wu Y, Wu Y, Hui T, Wu H, Wu Y, Wang W. Reaper homologue IBM1 in silkwormBombyx moriinduces apoptosis upon baculovirus infection. FEBS Lett 2013; 587:600-6. [DOI: 10.1016/j.febslet.2013.01.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/16/2013] [Accepted: 01/31/2013] [Indexed: 01/24/2023]
|
222
|
Krepler C, Chunduru SK, Halloran MB, He X, Xiao M, Vultur A, Villanueva J, Mitsuuchi Y, Neiman EM, Benetatos C, Nathanson KL, Amaravadi RK, Pehamberger H, McKinlay M, Herlyn M. The novel SMAC mimetic birinapant exhibits potent activity against human melanoma cells. Clin Cancer Res 2013; 19:1784-94. [PMID: 23403634 DOI: 10.1158/1078-0432.ccr-12-2518] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Inhibitor of apoptosis proteins (IAP) promote cancer cell survival and confer resistance to therapy. We report on the ability of second mitochondria-derived activator of caspases mimetic, birinapant, which acts as antagonist to cIAP1 and cIAP2, to restore the sensitivity to apoptotic stimuli such as TNF-α in melanomas. EXPERIMENTAL DESIGN Seventeen melanoma cell lines, representing five major genetic subgroups of cutaneous melanoma, were treated with birinapant as a single agent or in combination with TNF-α. Effects on cell viability, target inhibition, and initiation of apoptosis were assessed and findings were validated in 2-dimensional (2D), 3D spheroid, and in vivo xenograft models. RESULTS When birinapant was combined with TNF-α, strong combination activity, that is, neither compound was effective individually but the combination was highly effective, was observed in 12 of 18 cell lines. This response was conserved in spheroid models, whereas in vivo birinapant inhibited tumor growth without adding TNF-α in in vitro resistant cell lines. Birinapant combined with TNF-α inhibited the growth of a melanoma cell line with acquired resistance to BRAF inhibition to the same extent as in the parental cell line. CONCLUSIONS Birinapant in combination with TNF-α exhibits a strong antimelanoma effect in vitro. Birinapant as a single agent shows in vivo antitumor activity, even if cells are resistant to single agent therapy in vitro. Birinapant in combination with TNF-α is effective in a melanoma cell line with acquired resistance to BRAF inhibitors.
Collapse
Affiliation(s)
- Clemens Krepler
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Silke J, Meier P. Inhibitor of apoptosis (IAP) proteins-modulators of cell death and inflammation. Cold Spring Harb Perspect Biol 2013; 5:5/2/a008730. [PMID: 23378585 DOI: 10.1101/cshperspect.a008730] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Misregulated innate immune signaling and cell death form the basis of much human disease pathogenesis. Inhibitor of apoptosis (IAP) protein family members are frequently overexpressed in cancer and contribute to tumor cell survival, chemo-resistance, disease progression, and poor prognosis. Although best known for their ability to regulate caspases, IAPs also influence ubiquitin (Ub)-dependent pathways that modulate innate immune signaling via activation of nuclear factor κB (NF-κB). Recent research into IAP biology has unearthed unexpected roles for this group of proteins. In addition, the advances in our understanding of the molecular mechanisms that IAPs use to regulate cell death and innate immune responses have provided new insights into disease states and suggested novel intervention strategies. Here we review the functions assigned to those IAP proteins that act at the intersection of cell death regulation and inflammatory signaling.
Collapse
Affiliation(s)
- John Silke
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.
| | | |
Collapse
|
224
|
Hashimoto K, Saito B, Miyamoto N, Oguro Y, Tomita D, Shiokawa Z, Asano M, Kakei H, Taya N, Kawasaki M, Sumi H, Yabuki M, Iwai K, Yoshida S, Yoshimatsu M, Aoyama K, Kosugi Y, Kojima T, Morishita N, Dougan DR, Snell GP, Imamura S, Ishikawa T. Design and Synthesis of Potent Inhibitor of Apoptosis (IAP) Proteins Antagonists Bearing an Octahydropyrrolo[1,2-a]pyrazine Scaffold as a Novel Proline Mimetic. J Med Chem 2013; 56:1228-46. [DOI: 10.1021/jm301674z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kentaro Hashimoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Bunnai Saito
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naoki Miyamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuya Oguro
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Daisuke Tomita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Zenyu Shiokawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Moriteru Asano
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroyuki Kakei
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naohiro Taya
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masanori Kawasaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroyuki Sumi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masato Yabuki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kenichi Iwai
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Sei Yoshida
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mie Yoshimatsu
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazunobu Aoyama
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yohei Kosugi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takashi Kojima
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Nao Morishita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Douglas R. Dougan
- Structural Biology, Takeda California, Inc., 10410 Science Center Drive,
San Diego, California 92121, United States
| | - Gyorgy P. Snell
- Structural Biology, Takeda California, Inc., 10410 Science Center Drive,
San Diego, California 92121, United States
| | - Shinichi Imamura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomoyasu Ishikawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi
2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
225
|
Chen F, Xu C, Du L, Wang Y, Cao J, Fu Y, Guo Y, Liu Q, Fan F. Tat-SmacN7 induces radiosensitization in cancer cells through the activation of caspases and induction of apoptosis. Int J Oncol 2013; 42:985-92. [PMID: 23338568 DOI: 10.3892/ijo.2013.1785] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/21/2012] [Indexed: 11/05/2022] Open
Abstract
A major concern in cancer therapy is resistance of tumors such as human non-small cell lung cancer and esophageal cancer to radiotherapy. Intrinsic radioresistance of these cancer cells limits therapeutic efficiency. Here, we determined in two cancer cell lines the potential radiosensitizing activity of Tat-SmacN7, a small molecule compound, which mimics the activity of Smac, a mitochondrial protein released during apoptosis. We found that Tat-SmacN7 can enter the cells and promote RNA expression and the activity of caspase-3, -8 and -9 and sensitized the cancer cells to radiation with a sensitization enhancement ratio (SER) of 1.5-1.6. Tat-SmacN7 radiosensitization was mediated by both extrinsic and intrinsic apoptosis pathways through activation of caspases. Consistently, blockage of caspase activation, through treatment with a caspase inhibitor, z-VAD-fmk, inhibited apoptosis and abrogated Tat-SmacN7 radiosensitization. Our study demonstrates that Tat-SmacN7 also has radiosensitization effects in vivo, so it could be further developed as a novel class of radiosensitizers for the treatment of radioresistant human non-small cell lung cancer and esophageal cancer.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Radiation Hazard Evaluation, Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Winkler J, Rand ML, Schmugge M, Speer O. Omi/HtrA2 and XIAP are components of platelet apoptosis signalling. Thromb Haemost 2013; 109:532-9. [PMID: 23306356 DOI: 10.1160/th12-06-0404] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 12/05/2012] [Indexed: 12/18/2022]
Abstract
Although platelets possess the hallmarks of apoptosis such as activation of caspases, cytochrome c release and depolarisation of the mitochondrial transmembrane potential (∆Ψm), their entire apoptotic-signalling pathway is not totally understood. Therefore we studied the expression of various apoptotic proteins and found that platelets contain the pro-apoptotic proteins Omi/HtrA2 and Smac/Diablo, as well as their target the X-linked inhibitor of apoptosis XIAP. Omi/HtrA2 and Smac/Diablo were released from mitochondria into the platelet cytosol together with cytochrome c after induction of apoptosis by the Ca2+ ionophore A23187 or the BH3 mimetic ABT-737, and to a lesser extent, after platelet stimulation with collagen and thrombin. Inhibition of Omi/HtrA2 led to decreased levels of activated caspase-3/7 and caspase-9, but did not abolish loss of ∆Ψm or prevent release of Omi/HtrA2 from mitochondria. These results indicate that platelets have a functional intrinsic apoptotic-signalling pathway including the pro-apoptotic protease Omi/HtrA2 and its target protein XIAP.
Collapse
Affiliation(s)
- Jeannine Winkler
- Division of Haematology and Children's Research Center, University Children's Hospital Zurich, and Zurich Center for Integrative Human Physiology, University of Zurich, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland
| | | | | | | |
Collapse
|
227
|
Wang K, Lin B. Pathophysiological Significance of Hepatic Apoptosis. ISRN HEPATOLOGY 2012; 2013:740149. [PMID: 27335822 PMCID: PMC4890876 DOI: 10.1155/2013/740149] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 12/13/2012] [Indexed: 12/19/2022]
Abstract
Apoptosis is a classical pathological feature in liver diseases caused by various etiological factors such as drugs, viruses, alcohol, and cholestasis. Hepatic apoptosis and its deleterious effects exacerbate liver function as well as involvement in fibrosis/cirrhosis and carcinogenesis. An imbalance between apoptotic and antiapoptotic capabilities is a prominent characteristic of liver injury. The regulation of apoptosis and antiapoptosis can be a pivotal step in the treatment of liver diseases.
Collapse
Affiliation(s)
- Kewei Wang
- Departments of Surgery and Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Bingliang Lin
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
228
|
Qin S, Yang C, Wang X, Xu C, Li S, Zhang B, Ren H. Overexpression of Smac promotes Cisplatin-induced apoptosis by activating caspase-3 and caspase-9 in lung cancer A549 cells. Cancer Biother Radiopharm 2012; 28:177-82. [PMID: 23252748 DOI: 10.1089/cbr.2012.1261] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Second mitochondrial-derived activator of caspase (Smac) plays crucial roles in mitochondrial apoptosis pathways and promotes chemotherapy-induced apoptosis. In this study, Smac levels were examined in various lung cancer cell lines, and the effects of overexpressed Smac in the nonsmall-cell lung cancer cell line A549 were assayed by stable transfection of Smac. Subsequently, MTT assays, cell counting, and flow cytometry were applied to show that overexpression of Smac inhibits cell viability and cell growth and enhances apoptosis after cisplatin treatment. Western blotting was performed before and after cisplatin treatment to demonstrate that drug treatment could release Smac from mitochondria into the cytosol and promote apoptosis by activating caspase-3 and caspase-9. Promotion of apoptosis by cytosolic Smac could be blocked by pretreating cells with the caspase-9 inhibitor z-LEHD-FMK. Our findings indicate that overexpressed Smac significantly inhibited A549 cell growth and promoted apoptosis following cisplatin treatment due to the release of Smac from mitochondria into the cytosol, which increased the activities of caspase-3 and caspase-9.
Collapse
Affiliation(s)
- Sida Qin
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| | | | | | | | | | | | | |
Collapse
|
229
|
Sumi H, Yabuki M, Iwai K, Morimoto M, Hibino R, Inazuka M, Hashimoto K, Kosugi Y, Aoyama K, Yamamoto S, Yoshimatsu M, Yamasaki H, Tozawa R, Ishikawa T, Yoshida S. Antitumor Activity and Pharmacodynamic Biomarkers of a Novel and Orally Available Small-Molecule Antagonist of Inhibitor of Apoptosis Proteins. Mol Cancer Ther 2012; 12:230-40. [DOI: 10.1158/1535-7163.mct-12-0699] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
230
|
Blockade of inhibitors of apoptosis (IAPs) in combination with tumor-targeted delivery of tumor necrosis factor-α leads to synergistic antitumor activity. Cancer Gene Ther 2012; 20:46-56. [PMID: 23154431 PMCID: PMC3534156 DOI: 10.1038/cgt.2012.83] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the current study, we examined whether the combination of tumor vasculature-targeted gene therapy with adeno-associated virus bacteriophage-tumor necrosis factor-α (AAVP-TNF-α) and/or the orally administered LCL161, an antagonist of inhibitors of apoptosis proteins (IAPs), enhanced antitumor efficacy without systemic toxicity. M21 human melanoma xenografts were grown subcutaneously in nude mice. Mice were treated according to one of four treatment regimens: AAVP-TNF-α alone (AAVP-TNF-α plus sodium acetate-acetic acid (NaAc) buffer) via tail vein injection; LCL161 alone (phosphate-buffered saline (PBS) plus LCL161) via oral gavage; AAVP-TNF-α plus LCL161; and PBS plus NaAc Buffer as a control group. Tumor volume, survival and toxicity were analyzed. AAVP trafficking and TNF-α production in vivo were detected on days 7 and 21 by real-time PCR, enzyme-linked immunosorbent assay and immunofluorescence. The levels of apoptosis and activation of caspases were assessed on days 7 and 21 by TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling) and immunofluorescence assays. Our results showed that the combination of AAVP-TNF-α and LCL161 significantly inhibited tumor growth and prolonged survival in mice with melanoma xenografts. The combination of AAVP-TNF-α and LCL161 was also significantly more effective than either agent alone, showing a synergistic effect without systemic toxicity.
Collapse
|
231
|
Cossu F, Milani M, Vachette P, Malvezzi F, Grassi S, Lecis D, Delia D, Drago C, Seneci P, Bolognesi M, Mastrangelo E. Structural insight into inhibitor of apoptosis proteins recognition by a potent divalent smac-mimetic. PLoS One 2012; 7:e49527. [PMID: 23166698 PMCID: PMC3499469 DOI: 10.1371/journal.pone.0049527] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/10/2012] [Indexed: 11/18/2022] Open
Abstract
Genetic alterations enhancing cell survival and suppressing apoptosis are hallmarks of cancer that significantly reduce the efficacy of chemotherapy or radiotherapy. The Inhibitor of Apoptosis Protein (IAP) family hosts conserved proteins in the apoptotic pathway whose over-expression, frequently found in tumours, potentiates survival and resistance to anticancer agents. In humans, IAPs comprise eight members hosting one or more structural Baculoviral IAP Repeat (BIR) domains. Cellular IAPs (cIAP1 and 2) indirectly inhibit caspase-8 activation, and regulate both the canonical and the non-canonical NF-κB signaling pathways. In contrast to cIAPs, XIAP (X chromosome-linked Inhibitor of Apoptosis Protein) inhibits directly the effector caspases-3 and -7 through its BIR2 domain, and initiator caspase-9 through its BIR3 domain; molecular docking studies suggested that Smac/DIABLO antagonizes XIAP by simultaneously targeting both BIR2 and BIR3 domains. Here we report analytical gel filtration, crystallographic and SAXS experiments on cIAP1-BIR3, XIAP-BIR3 and XIAP-BIR2BIR3 domains, alone and in the presence of compound 9a, a divalent homodimeric Smac mimetic. 9a is shown to bind two BIR domains inter- (in the case of two BIR3) and intra-molecularly (in the case of XIAP-BIR2BIR3), with higher affinity for cIAP1-BIR3, relative to XIAP-BIR3. Despite the different crystal lattice packing, 9a maintains a right handed helical conformation in both cIAP1-BIR3 and XIAP-BIR3 crystals, that is likely conserved in solution as shown by SAXS data. Our structural results demonstrate that the 9a linker length, its conformational degrees of freedom and its hydrophobicity, warrant an overall compact structure with optimal solvent exposure of its two active moieties for IAPs binding. Our results show that 9a is a good candidate for pre-clinical and clinical studies, worth of further investigations in the field of cancer therapy.
Collapse
Affiliation(s)
- Federica Cossu
- Department of Biosciences, University of Milano, Milano, Italy
| | - Mario Milani
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
| | - Patrice Vachette
- Institut de Biochimie et de Biophysique Moléculaire et Cellulaire, UMR8619 CNRS, Université Paris-Sud, IFR115, Orsay, France
| | | | - Serena Grassi
- Department of Biosciences, University of Milano, Milano, Italy
| | | | | | - Carmelo Drago
- Centro Interdisciplinare Studi bio-molecolari e applicazioni Industriali (CISI), University of Milano, Milano, Italy
| | - Pierfausto Seneci
- Centro Interdisciplinare Studi bio-molecolari e applicazioni Industriali (CISI), University of Milano, Milano, Italy
- Department of Organic and Industrial Chemistry, University of Milano, Milano, Italy
| | - Martino Bolognesi
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
| | - Eloise Mastrangelo
- Department of Biosciences, University of Milano, Milano, Italy
- CNR-Istituto di Biofisica, Università degli Studi di Milano, Milano, Italy
- * E-mail:
| |
Collapse
|
232
|
The role of the IAP E3 ubiquitin ligases in regulating pattern-recognition receptor signalling. Nat Rev Immunol 2012; 12:833-44. [PMID: 23124073 DOI: 10.1038/nri3325] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An inflammatory response is initiated when innate immune pattern-recognition receptors (PRRs) expressed by different cell types detect constituents of invading microorganisms and endogenous intracellular molecules released by dying cells. The intracellular cascades activated by PRRs induce the expression and maturation of inflammatory molecules that coordinate the removal of the infectious agents and of the infected or damaged cells. In this Review, we discuss the findings implicating members of the inhibitor of apoptosis protein (IAP) family in the ubiquitylation-dependent regulation of PRR signalling. Understanding the role of IAPs in innate immunity may open new therapeutic perspectives for the treatment of PRR-dependent inflammatory diseases.
Collapse
|
233
|
Manzoni L, Belvisi L, Bianchi A, Conti A, Drago C, de Matteo M, Ferrante L, Mastrangelo E, Perego P, Potenza D, Scolastico C, Servida F, Timpano G, Vasile F, Rizzo V, Seneci P. Homo- and heterodimeric Smac mimetics/IAP inhibitors as in vivo-active pro-apoptotic agents. Part I: Synthesis. Bioorg Med Chem 2012; 20:6687-708. [DOI: 10.1016/j.bmc.2012.09.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 12/22/2022]
|
234
|
Marivin A, Berthelet J, Plenchette S, Dubrez L. The Inhibitor of Apoptosis (IAPs) in Adaptive Response to Cellular Stress. Cells 2012; 1:711-37. [PMID: 24710527 PMCID: PMC3901146 DOI: 10.3390/cells1040711] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/14/2012] [Accepted: 09/27/2012] [Indexed: 12/31/2022] Open
Abstract
Cells are constantly exposed to endogenous and exogenous cellular injuries. They cope with stressful stimuli by adapting their metabolism and activating various "guardian molecules." These pro-survival factors protect essential cell constituents, prevent cell death, and possibly repair cellular damages. The Inhibitor of Apoptosis (IAPs) proteins display both anti-apoptotic and pro-survival properties and their expression can be induced by a variety of cellular stress such as hypoxia, endoplasmic reticular stress and DNA damage. Thus, IAPs can confer tolerance to cellular stress. This review presents the anti-apoptotic and survival functions of IAPs and their role in the adaptive response to cellular stress. The involvement of IAPs in human physiology and diseases in connection with a breakdown of cellular homeostasis will be discussed.
Collapse
Affiliation(s)
- Arthur Marivin
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR866, Dijon F-21079, France.
| | - Jean Berthelet
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR866, Dijon F-21079, France.
| | - Stéphanie Plenchette
- Institut Fédératif de Recherche (IFR), Université de Bourgogne, 100, Dijon F-21079, France.
| | - Laurence Dubrez
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR866, Dijon F-21079, France.
| |
Collapse
|
235
|
Bas E, Dinh CT, Garnham C, Polak M, Van de Water TR. Conservation of hearing and protection of hair cells in cochlear implant patients' with residual hearing. Anat Rec (Hoboken) 2012; 295:1909-27. [DOI: 10.1002/ar.22574] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/13/2022]
|
236
|
Thompson AD, Dugan A, Gestwicki JE, Mapp AK. Fine-tuning multiprotein complexes using small molecules. ACS Chem Biol 2012; 7:1311-20. [PMID: 22725693 DOI: 10.1021/cb300255p] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Multiprotein complexes such as the transcriptional machinery, signaling hubs, and protein folding machines are typically composed of at least one enzyme combined with multiple non-enzymes. Often the components of these complexes are incorporated in a combinatorial manner, in which the ultimate composition of the system helps dictate the type, location, or duration of cellular activities. Although drugs and chemical probes have traditionally targeted the enzyme components, emerging strategies call for controlling the function of protein complexes by modulation of protein-protein interactions (PPIs). However, the challenges of targeting PPIs have been well documented, and the diversity of PPIs makes a "one-size-fits-all" solution highly unlikely. These hurdles are particularly daunting for PPIs that encompass large buried surface areas and those with weak affinities. In this Review, we discuss lessons from natural systems, in which allostery and other mechanisms are used to overcome the challenge of regulating the most difficult PPIs. These systems may provide a blueprint for identifying small molecules that target challenging PPIs and affecting molecular decision-making within multiprotein systems.
Collapse
Affiliation(s)
- Andrea D. Thompson
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amanda Dugan
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jason E. Gestwicki
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Anna K. Mapp
- Program
in Chemical Biology, ‡Departments of Pathology and Biological Chemistry and the Life Sciences
Institute, and §Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
237
|
Abstract
Mitochondria have been classically characterized as organelles with responsibility for cellular energy production in the form of ATP, but they are also the organelles through which apoptotic signaling occurs. Cell stress stimuli can result in outer membrane permeabilization, after which mitochondria release numerous proteins involved in apoptotic signaling, including cytochrome c, apoptosis-inducing factor, endonuclease G, Smac/DIABLO and Omi/HtrA2. Cell fate is determined by signaling through apoptotic proteins within the Bcl-2 (B-cell lymphoma 2) protein family, which converges on mitochondria. Many cancerous cells display abnormal levels of Bcl-2 protein family member expression that results in defective apoptotic signaling. Alterations in bioenergetic function also contribute to cancer as well as numerous other disorders. Recent evidence indicates that several pro-apoptotic proteins localized within mitochondria, as well as proteins within the Bcl-2 protein family, can influence mitochondrial bioenergetic function. This review focuses on the emerging roles of these proteins in the control of mitochondrial activity.
Collapse
Affiliation(s)
- S M Kilbride
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | |
Collapse
|
238
|
Phillipps HR, Hurst PR. XIAP: a potential determinant of ovarian follicular fate. Reproduction 2012; 144:165-76. [DOI: 10.1530/rep-12-0142] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
X-linked inhibitor of apoptosis protein (XIAP), a member of the inhibitor of apoptosis protein family, is involved in regulating a number of functions including receptor-mediated intracellular signalling and early development. Its role as an endogenous caspase inhibitor, however, is the most highly characterised. Consequently, this protein has been implicated as an anti-apoptotic factor in the ovary.In vitroandin vivostudies have begun dissecting the stimuli and signalling networks that lead to XIAP upregulation in granulosa cells. The objective of this review is to briefly summarise the current knowledge concerning XIAP and its interactions with different caspases. Furthermore, XIAP's emerging role in the mammalian ovary will be explored and comparison is made with its functions in the mammary gland. Finally, the idea that XIAP may act as a molecular signalling switch in granulosa cells following detachment from underlying layers to promote follicular atresia will be introduced.
Collapse
|
239
|
Foster DM, Stauffer SH, Stone MR, Gookin JL. Proteasome inhibition of pathologic shedding of enterocytes to defend barrier function requires X-linked inhibitor of apoptosis protein and nuclear factor κB. Gastroenterology 2012; 143:133-44.e4. [PMID: 22446197 DOI: 10.1053/j.gastro.2012.03.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 03/07/2012] [Accepted: 03/12/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Although we are beginning to understand where, when, and how intestinal epithelial cells are shed, physiologically, less is understood about alterations in cell fate during minimally invasive epithelial infections. We used a piglet model of Cryptosporidium parvum infection to determine how elimination of infected enterocytes is balanced with the need to maintain barrier function. METHODS We studied the effects of enterocyte shedding by C parvum-infected ileum on barrier function ex vivo with Ussing chambers. The locations and activities of caspase-3, nuclear factor κB (NF-κB), and inhibitor of apoptosis proteins (IAP) were assayed by enzyme-linked immunosorbent assay, immunoblot, and tissue immunoreactivity analyses and using specific pharmacologic inhibitors. The location, specificity, and magnitude of enterocyte shedding were quantified using special stains and light microscopy. RESULTS Infection with C parvum activated apoptotic signaling pathways in enterocytes that resulted in cleavage of caspase-3. Despite caspase-3 cleavage, enterocyte shedding was confined to villus tips, coincident with apoptosis, and observed more frequently in infected cells. Epithelial expression of X-linked inhibitor of apoptosis protein (XIAP), activation of NF-κB, and proteasome activity were required for control of cell shedding and barrier function. The proteasome blocked activity of caspase-3; this process was mediated by expression of XIAP, which bound to cleaved caspase-3. CONCLUSIONS We have identified a pathway by which villus epithelial cells are maintained during C parvum infection. Loss of barrier function is reduced by active retention of infected enterocytes until they reach the villus tip. These findings might be used to promote clearance of minimally invasive enteropathogens, such as by increasing the rate of migration of epithelial cells from the crypt to the villus tip.
Collapse
Affiliation(s)
- Derek M Foster
- Department of Population Health, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27607, USA
| | | | | | | |
Collapse
|
240
|
Wynendaele E, Pauwels E, Van de Wiele C, Burvenich C, De Spiegeleer B. The potential role of quorum-sensing peptides in oncology. Med Hypotheses 2012; 78:814-7. [DOI: 10.1016/j.mehy.2012.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 03/09/2012] [Accepted: 03/22/2012] [Indexed: 10/28/2022]
|
241
|
Ramachandiran S, Cain J, Liao A, He Y, Guo X, Boise LH, Fu H, Ratner L, Khoury HJ, Bernal-Mizrachi L. The Smac mimetic RMT5265.2HCL induces apoptosis in EBV and HTLV-I associated lymphoma cells by inhibiting XIAP and promoting the mitochondrial release of cytochrome C and Smac. Leuk Res 2012; 36:784-90. [PMID: 22325366 PMCID: PMC3331941 DOI: 10.1016/j.leukres.2011.12.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/29/2011] [Accepted: 12/30/2011] [Indexed: 12/14/2022]
Abstract
The inhibitors of apoptosis (IAP) are important regulators of apoptosis. However, little is known about the capacity of Smac mimetics (IAP inhibitor) to overcome virally associated-lymphoma's (VAL) resistance to apoptosis. Here, we explored the pro-apoptotic effect of a novel Smac mimetic, RMT5265.2HCL (RMT) in VAL cells. RMT improved the sensitivity to apoptosis in EBV- and to some extend in HTLV-1- but not in HHV-8-VAL. Furthermore, we identified that RMT promotes caspase 3 and 9 cleavage by inhibiting XIAP and inducing the mitochondrial efflux of Smac and cytochrome C. This investigation further support exploring the use of Smac inhibitors in VAL.
Collapse
Affiliation(s)
- Sampath Ramachandiran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Joan Cain
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Albert Liao
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yanjuan He
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Xiangxue Guo
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Haian Fu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Pharmacology, Emory University, Atlanta, GA, USA
| | - Lee Ratner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hanna Jean Khoury
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Leon Bernal-Mizrachi
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
242
|
Du J, Kelly AE, Funabiki H, Patel DJ. Structural basis for recognition of H3T3ph and Smac/DIABLO N-terminal peptides by human Survivin. Structure 2012; 20:185-95. [PMID: 22244766 DOI: 10.1016/j.str.2011.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 11/29/2011] [Accepted: 12/02/2011] [Indexed: 11/18/2022]
Abstract
Survivin is an inhibitor of apoptosis family protein implicated in apoptosis and mitosis. In apoptosis, it has been shown to recognize the Smac/DIABLO protein. It is also a component of the chromosomal passenger complex, a key player during mitosis. Recently, Survivin was identified in vitro and in vivo as the direct binding partner for phosphorylated Thr3 on histone H3 (H3T3ph). We have undertaken structural and binding studies to investigate the molecular basis underlying recognition of H3T3ph and Smac/DIABLO N-terminal peptides by Survivin. Our crystallographic studies establish recognition of N-terminal Ala in both complexes and identify intermolecular hydrogen-bonding interactions in the Survivin phosphate-binding pocket that contribute to H3T3ph mark recognition. In addition, our calorimetric data establish that Survivin binds tighter to the H3T3ph-containing peptide relative to the N-terminal Smac/DIABLO peptide, and this preference can be reversed through structure-guided mutations that increase the hydrophobicity of the phosphate-binding pocket.
Collapse
Affiliation(s)
- Jiamu Du
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | |
Collapse
|
243
|
Masoumi KC, Cornmark L, Lønne GK, Hellman U, Larsson C. Identification of a novel protein kinase Cδ-Smac complex that dissociates during paclitaxel-induced cell death. FEBS Lett 2012; 586:1166-72. [PMID: 22465666 DOI: 10.1016/j.febslet.2012.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 02/23/2012] [Accepted: 03/05/2012] [Indexed: 11/25/2022]
Abstract
Protein kinase C (PKC) δ is a regulator of apoptosis with both pro- and anti-apoptotic effects. The mechanistic basis for the discrepant effects is not completely understood. Here we show that Smac interacts with PKCδ. The interaction depends on the N-terminus of Smac and is disrupted upon treatment with paclitaxel. This is associated with release of Smac into the cytosol. Activation of PKCδ rescues the interaction during paclitaxel exposure and suppresses the paclitaxel-mediated cell death. However, under these conditions the complex is mainly found in the cytosol suggesting that cytosolic Smac can be bound by PKCδ when PKC is activated. The data unravel a previously unrecognized interaction and suggest that PKCδ by associating with Smac may prevent its apoptotic effects.
Collapse
|
244
|
He X, Khurana A, Maguire JL, Chien J, Shridhar V. HtrA1 sensitizes ovarian cancer cells to cisplatin-induced cytotoxicity by targeting XIAP for degradation. Int J Cancer 2012; 130:1029-35. [PMID: 21387310 PMCID: PMC3206182 DOI: 10.1002/ijc.26044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 02/22/2011] [Indexed: 01/13/2023]
Abstract
HtrA1, a member of serine protease family, has been previously found to be involved in resistance to chemotherapy in ovarian cancer although the underlying mechanism is not clear. Using mixture-based oriented peptide library approach, previously we identified X-linked inhibitor of apoptosis protein (XIAP), a member of the inhibitor of apoptosis proteins family, as a potential substrate of HtrA1. The aim of our work is to investigate the link between HtrA1 and XIAP proteins and their relationships with chemoresistance in ovarian cancer. Our results showed that recombinant XIAP was degraded by purified wild-type HtrA1 but not mutant HtrA1 in vitro. Consistent with the in vitro data, coimmunoprecipitation assays showed that HtrA1 and XIAP formed a protein complex in vivo. Ectopic expression of HtrA1 led to decreased level of XIAP in OV167 and OV202 ovarian cancer cells, while knockdown of HtrA1 resulted in increased level of XIAP in SKOV3 ovarian cancer cells. Furthermore, overexpression of HtrA1 in OV202 cells promoted cell sensitivity to cisplatin-induced apoptosis that could be reversed by increased expression of XIAP. The cleavage of XIAP induced by HtrA1 was enhanced by cisplatin treatment. Taken together, our experiments have identified XIAP as a novel substrate of HtrA1 and the degradation of XIAP by HtrA1 contributes to cell response to chemotherapy, suggesting that restoring the expression of HtrA1 may be a promising treatment strategy for ovarian cancer.
Collapse
Affiliation(s)
- Xiaoping He
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
245
|
Feltham R, Khan N, Silke J. IAPS and ubiquitylation. IUBMB Life 2012; 64:411-8. [DOI: 10.1002/iub.565] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/25/2011] [Indexed: 11/11/2022]
|
246
|
Bianchi A, Ugazzi M, Ferrante L, Lecis D, Scavullo C, Mastrangelo E, Seneci P. Rational design, synthesis and characterization of potent, drug-like monomeric Smac mimetics as pro-apoptotic anticancer agents. Bioorg Med Chem Lett 2012; 22:2204-8. [PMID: 22342627 DOI: 10.1016/j.bmcl.2012.01.098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 11/15/2022]
Abstract
A set of phenyl-substituted Smac mimetics/IAP inhibitor analogues of lead compound 2a was synthesized, aiming to retain its strong cell-free potency while increasing its bioavailability. Seventeen compounds 2b-r were prepared and characterized in vitro, using cell-free and cellular assays. Among them, the p-CF(3) substituted analogue 2m showed the best permeability through cell membranes, and was selected for further in vitro and in vivo studies due to its strong, sub-micromolar cellular potency.
Collapse
Affiliation(s)
- Aldo Bianchi
- CISI scrl, Via Fantoli 16/15, I-20138 Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
247
|
Fulda S, Vucic D. Targeting IAP proteins for therapeutic intervention in cancer. Nat Rev Drug Discov 2012; 11:109-24. [PMID: 22293567 DOI: 10.1038/nrd3627] [Citation(s) in RCA: 641] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Evasion of apoptosis is one of the crucial acquired capabilities used by cancer cells to fend off anticancer therapies. Inhibitor of apoptosis (IAP) proteins exert a range of biological activities that promote cancer cell survival and proliferation. X chromosome-linked IAP is a direct inhibitor of caspases - pro-apoptotic executioner proteases - whereas cellular IAP proteins block the assembly of pro-apoptotic protein signalling complexes and mediate the expression of anti-apoptotic molecules. Furthermore, mutations, amplifications and chromosomal translocations of IAP genes are associated with various malignancies. Among the therapeutic strategies that have been designed to target IAP proteins, the most widely used approach is based on mimicking the IAP-binding motif of second mitochondria-derived activator of caspase (SMAC), which functions as an endogenous IAP antagonist. Alternative strategies include transcriptional repression and the use of antisense oligonucleotides. This Review provides an update on IAP protein biology as well as current and future perspectives on targeting IAP proteins for therapeutic intervention in human malignancies.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstr. 3a, 60528 Frankfurt, Germany.
| | | |
Collapse
|
248
|
Smac: Its role in apoptosis induction and use in lung cancer diagnosis and treatment. Cancer Lett 2012; 318:9-13. [PMID: 22227574 DOI: 10.1016/j.canlet.2011.12.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 12/11/2011] [Accepted: 12/13/2011] [Indexed: 01/23/2023]
Abstract
Apoptosis is a conserved and regulated cell suicide process, the malfunction of which is closely linked with carcinogenesis. Caspases control the induction of apoptosis through an enzymatic cascade that can be activated by both the mitochondrial and death receptor pathways. Smac is a mitochondrial protein that interacts with Inhibitor of Apoptosis Proteins (IAPs) and, upon apoptotic stimuli, is released into the cytoplasm to inhibit the capase-binding activity of IAPs. Smac plays key roles in both the diagnosis and treatment of cancer, especially lung cancer. Our review will focus on the roles of Smac in lung carcinogenesis and cancer progression and its relevance in lung cancer treatment.
Collapse
|
249
|
Potenza D, Belvisi L, Vasile F, Moroni E, Cossu F, Seneci P. A NMR and computational study of Smac mimics targeting both the BIR2 and BIR3 domains in XIAP protein. Org Biomol Chem 2012; 10:3278-87. [DOI: 10.1039/c2ob06979b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
250
|
|