51
|
Vigneswara V, Ahmed Z. The Role of Caspase-2 in Regulating Cell Fate. Cells 2020; 9:cells9051259. [PMID: 32438737 PMCID: PMC7290664 DOI: 10.3390/cells9051259] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Caspase-2 is the most evolutionarily conserved member of the mammalian caspase family and has been implicated in both apoptotic and non-apoptotic signaling pathways, including tumor suppression, cell cycle regulation, and DNA repair. A myriad of signaling molecules is associated with the tight regulation of caspase-2 to mediate multiple cellular processes far beyond apoptotic cell death. This review provides a comprehensive overview of the literature pertaining to possible sophisticated molecular mechanisms underlying the multifaceted process of caspase-2 activation and to highlight its interplay between factors that promote or suppress apoptosis in a complicated regulatory network that determines the fate of a cell from its birth and throughout its life.
Collapse
|
52
|
Liu R, Wang X, Curtiss C, Sheikh MS, Huang Y. Monoglyceride lipase mediates tumor-suppressive effects by promoting degradation of X-linked inhibitor of apoptosis protein. Cell Death Differ 2020; 27:2888-2903. [PMID: 32376875 DOI: 10.1038/s41418-020-0549-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 11/09/2022] Open
Abstract
We have previously reported that Monoglyceride Lipase (MGL) expression is absent or reduced in various human malignancies and MGL-deficient mice develop tumors in multiple organs. Evidence also suggests MGL to be a tumor suppressor, however, the mechanisms underlying its tumor-suppressive actions remain to be investigated. Here, we report a novel function of MGL as a negative regulator of XIAP, an important inhibitor of apoptosis. We found that MGL directly interacted with XIAP and enhanced E3-ligase activity and proteasomal degradation of XIAP. MGL overexpression induced cell death that was coupled with caspase activation and reduced XIAP levels. N-terminus of MGL was found to mediate interactions with XIAP and induce cell death. MGL-deficient cells exhibited elevated XIAP levels and exhibited resistance to anticancer drugs. XIAP expression was significantly elevated in tissues of MGL-deficient animals as well as human lung cancers exhibiting reduced MGL expression. Thus, MGL appears to mediate its tumor-suppressive actions by inhibiting XIAP to induce cell death.
Collapse
Affiliation(s)
- Renyan Liu
- Department of Pharmacology, Upstate Medical University State University of New York, Syracuse, NY, 13210, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Bostone, MA, 02215, USA
| | - Xin Wang
- Department of Pharmacology, Upstate Medical University State University of New York, Syracuse, NY, 13210, USA
| | - Christopher Curtiss
- Department of Pathology, Upstate Medical University State University of New York, Syracuse, NY, 13210, USA
| | - M Saeed Sheikh
- Department of Pharmacology, Upstate Medical University State University of New York, Syracuse, NY, 13210, USA
| | - Ying Huang
- Department of Pharmacology, Upstate Medical University State University of New York, Syracuse, NY, 13210, USA.
| |
Collapse
|
53
|
IAP-Mediated Protein Ubiquitination in Regulating Cell Signaling. Cells 2020; 9:cells9051118. [PMID: 32365919 PMCID: PMC7290580 DOI: 10.3390/cells9051118] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Over the last decade, the E3-ubiquitine ligases from IAP (Inhibitor of Apoptosis) family have emerged as potent regulators of immune response. In immune cells, they control signaling pathways driving differentiation and inflammation in response to stimulation of tumor necrosis factor receptor (TNFR) family, pattern-recognition receptors (PRRs), and some cytokine receptors. They are able to control the activity, the cellular fate, or the stability of actors of signaling pathways, acting at different levels from components of receptor-associated multiprotein complexes to signaling effectors and transcription factors, as well as cytoskeleton regulators. Much less is known about ubiquitination substrates involved in non-immune signaling pathways. This review aimed to present IAP ubiquitination substrates and the role of IAP-mediated ubiquitination in regulating signaling pathways.
Collapse
|
54
|
Xue C, Kang B, Su P, Wang D, Zhao F, Zhang J, Wang X, Lang H, Cao Z. MicroRNA-106b-5p participates in lead (Pb 2+)-induced cell viability inhibition by targeting XIAP in HT-22 and PC12 cells. Toxicol In Vitro 2020; 66:104876. [PMID: 32344020 DOI: 10.1016/j.tiv.2020.104876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/25/2022]
Abstract
Previous studies reported perturbed expressing of X-linked inhibitor of apoptosis protein (XIAP) under lead (Pb) exposure. However, researches on XIAP expression mainly focused on its transcriptional and post-translational regulation, rarely involving post-transcriptional mechanism manipulated by certain indispensable microRNAs (miRNAs). Interestingly, we unveiled that miR-106b-5p, a widely expressed miRNA in various tissues, is up-regulated by Pb2+-induced stress. Moreover, we found a binding site for miR-106b-5p in the 3'-UTR of xiap mRNA using bioinformatics analysis, and provided the evidences that miR-106b-5p can interact and function with this regulatory region via luciferase reporter assay. Our results further showed that miR-106b-5p down-regulates XIAP protein level, and suppression of miR-106b-5p reverses the decrease in both XIAP level and cell viability in Pb2+-treated HT-22 and PC12 cells. In brief, we identified a novel function of miR-106b-5p in the post-transcriptional regulation of XIAP expression associated with Pb neurotoxicity.
Collapse
Affiliation(s)
- Chong Xue
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China
| | - Beipei Kang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China; Department of Clinical Laboratory, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Peng Su
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China
| | - Diya Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China
| | - Fang Zhao
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China
| | - Jianbin Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China
| | - Xiaojing Wang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China; Department of Neurology and Endocrinology, 989 Hospital of PLA, Pingdingshan 467021, China
| | - Haiyang Lang
- School of Public Health, Air Force Military Medical University, Xi'an 710032, China.
| | - Zipeng Cao
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Military Medical University, Xi'an 710032, China.
| |
Collapse
|
55
|
Ge Y, Wang L, Li D, Zhao C, Li J, Liu T. Exploring the Extended Biological Functions of the Human Copper Chaperone of Superoxide Dismutase 1. Protein J 2020; 38:463-471. [PMID: 31140034 DOI: 10.1007/s10930-019-09824-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The human copper chaperone of SOD1 (designated as CCS) was discovered more than two decades ago. It is an important copper binding protein and a homolog of Saccharomyces cerevisiae LYS7. To date, no studies have systematically or specifically elaborated on the functional development of CCS. This review summarizes the essential information about CCS, such as its localization, 3D structure, and copper binding ability. An emphasis is placed on its interacting protein partners and its biological functions in vivo and in vitro. Three-dimensional structural analysis revealed that CCS is composed of three domains. Its primary molecular function is the delivery of copper to SOD1 and activation of SOD1. It has also been reported to bind to XIAP, Mia40, and X11α, and other proteins. Through these protein partners, CCS is implicated in several vital biological processes in vivo, such as copper homeostasis, apoptosis, angiogenesis and oxidative stress. This review is anticipated to assist scientists in systematically understanding the latest research developments of CCS for facilitating the development of new therapeutics targeting CCS in the future.
Collapse
Affiliation(s)
- Yan Ge
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Lu Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China. .,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China.
| | - Duanhua Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Chen Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Jinjun Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Tao Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 168 Huaguan Road, Chenghua District, Chengdu, 610052, China.,International Phage Drug Research Center, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| |
Collapse
|
56
|
Sandag Z, Jung S, Quynh NTN, Myagmarjav D, Anh NH, Le DDT, Lee BS, Mongre RK, Jo T, Lee M. Inhibitory Role of TRIP-Br1/XIAP in Necroptosis under Nutrient/Serum Starvation. Mol Cells 2020; 43:236-250. [PMID: 32050753 PMCID: PMC7103882 DOI: 10.14348/molcells.2020.2193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/19/2019] [Accepted: 01/02/2020] [Indexed: 12/16/2022] Open
Abstract
Currently, many available anti-cancer therapies are targeting apoptosis. However, many cancer cells have acquired resistance to apoptosis. To overcome this problem, simultaneous induction of other types of programmed cell death in addition to apoptosis of cancer cells might be an attractive strategy. For this purpose, we initially investigated the inhibitory role of TRIP-Br1/XIAP in necroptosis, a regulated form of necrosis, under nutrient/serum starvation. Our data showed that necroptosis was significantly induced in all tested 9 different types of cancer cell lines in response to prolonged serum starvation. Among them, necroptosis was induced at a relatively lower level in MCF-7 breast cancer line that was highly resistant to apoptosis than that in other cancer cell lines. Interestingly, TRIP-Br1 oncogenic protein level was found to be very high in this cell line. Upregulated TRIP-Br1 suppressed necroptosis by repressing reactive oxygen species generation. Such suppression of necroptosis was greatly enhanced by XIAP, a potent inhibitor of apoptosis. Our data also showed that TRIP-Br1 increased XIAP phosphorylation at serine87, an active form of XIAP. Our mitochondrial fractionation data revealed that TRIPBr1 protein level was greatly increased in the mitochondria upon serum starvation. It suppressed the export of CypD, a vital regulator in mitochondria-mediated necroptosis, from mitochondria to cytosol. TRIP-Br1 also suppressed shikoninmediated necroptosis, but not TNF-α-mediated necroptosis, implying possible presence of another signaling pathway in necroptosis. Taken together, our results suggest that TRIPBr1/XIAP can function as onco-proteins by suppressing necroptosis of cancer cells under nutrient/serum starvation.
Collapse
Affiliation(s)
- Zolzaya Sandag
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - Samil Jung
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | | | | | - Nguyen Hai Anh
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - Dan-Diem Thi Le
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - Beom Suk Lee
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - Raj Kumar Mongre
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - Taeyeon Jo
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| | - MyeongSok Lee
- Department of Biological Science, Sookmyung Women’s University, Seoul 430, Korea
| |
Collapse
|
57
|
Boice A, Bouchier-Hayes L. Targeting apoptotic caspases in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118688. [PMID: 32087180 DOI: 10.1016/j.bbamcr.2020.118688] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/20/2020] [Accepted: 02/15/2020] [Indexed: 12/30/2022]
Abstract
Members of the caspase family of proteases play essential roles in the initiation and execution of apoptosis. These caspases are divided into two groups: the initiator caspases (caspase-2, -8, -9 and -10), which are the first to be activated in response to a signal, and the executioner caspases (caspase-3, -6, and -7) that carry out the demolition phase of apoptosis. Many conventional cancer therapies induce apoptosis to remove the cancer cell by engaging these caspases indirectly. Newer therapeutic applications have been designed, including those that specifically activate individual caspases using gene therapy approaches and small molecules that repress natural inhibitors of caspases already present in the cell. For such approaches to have maximal clinical efficacy, emerging insights into non-apoptotic roles of these caspases need to be considered. This review will discuss the roles of caspases as safeguards against cancer in the context of the advantages and potential limitations of targeting apoptotic caspases for the treatment of cancer.
Collapse
Affiliation(s)
- Ashley Boice
- Department of Pediatrics, Division of Hematology-Oncology and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
58
|
Chen W, Song J, Bian H, Yang X, Xie X, Zhu Q, Qin C, Qi J. The functions and targets of miR-212 as a potential biomarker of cancer diagnosis and therapy. J Cell Mol Med 2020; 24:2392-2401. [PMID: 31930653 PMCID: PMC7028855 DOI: 10.1111/jcmm.14966] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 12/17/2019] [Accepted: 12/22/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer is a major health problem worldwide. An increasing number of researchers are studying the diagnosis, therapy and mechanisms underlying the development and progression of cancer. The study of noncoding RNA has attracted a lot of attention in recent years. It was found that frequent alterations of miRNA expression not only have various functions in cancer but also that miRNAs can act as clinical markers of diagnosis, stage and progression of cancer. MiR-212 is an important example of miRNAs involved in cancer. According to recent studies, miR-212 may serve as an oncogene or tumour suppressor by influencing different targets or pathways during the oncogenesis and the development and metastasis of cancer. Its deregulation may serve as a marker for the diagnosis or prognosis of cancer. In addition, it was recently reported that miR-212 was related to the sensitivity or resistance of cancer cells to chemotherapy or radiotherapy. Here, we summarize the current understanding of miR-212 functions in cancer by describing the relevant signalling pathways and targets. The role of miR-212 as a biomarker and its therapeutic potential in cancer is also described. The aim of this review was to identify new methods for the diagnosis and treatment of human cancers.
Collapse
Affiliation(s)
- Wenjun Chen
- Departments of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China.,Departments of Gastroenterology, The Affiliated Weihai Second Municipal Hospital of Qingdao University, Qingdao, China
| | - Jing Song
- Departments of Gastroenterology, The Affiliated Weihai Second Municipal Hospital of Qingdao University, Qingdao, China
| | - Hongjun Bian
- Departments of Emergency Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xia Yang
- Departments of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiaoyu Xie
- Departments of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Qiang Zhu
- Departments of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China
| | - Chengyong Qin
- Departments of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China
| | - Jianni Qi
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, China.,Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
59
|
Xu J, Hua X, Yang R, Jin H, Li J, Zhu J, Tian Z, Huang M, Jiang G, Huang H, Huang C. XIAP Interaction with E2F1 and Sp1 via its BIR2 and BIR3 domains specific activated MMP2 to promote bladder cancer invasion. Oncogenesis 2019; 8:71. [PMID: 31811115 PMCID: PMC6898186 DOI: 10.1038/s41389-019-0181-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 11/09/2022] Open
Abstract
XIAP has generally been thought to function in bladder cancer. However, the potential function of structure-based function of XIAP in human BC invasion has not been well explored before. We show here that ectopic expression of the BIR domains of XIAP specifically resulted in MMP2 activation and cell invasion in XIAP-deleted BC cells, while Src was further defined as an XIAP downstream negative regulator for MMP2 activation and BC cell invasion. The inhibition of Src expression by the BIR domains was caused by attenuation of Src protein translation upon miR-203 upregulation; which was resulted from direct interaction of BIR2 and BIR3 with E2F1 and Sp1, respectively. The interaction of BIR2/BIR3 with E2F1/Sp1 unexpectedly occurred, which could be blocked by serum-induced XIAP translocation. Taken together, our studies, for the first time revealed that: (1) BIR2 and BIR3 domains of XIAP play their role in cancer cell invasion without affecting cell migration by specific activation of MMP2 in human BC cells; (2) by BIR2 interacting with E2F1 and BIR3 interacting with Sp1, XIAP initiates E2F1/Sp1 positive feedback loop-dependent transcription of miR-203, which in turn inhibits Src protein translation, further leading to MMP2-cleaved activation; (3) XIAP interaction with E2F1 and Sp1 is observed in the nucleus. Our findings provide novel insights into understanding the specific function of BIR2 and BIR3 of XIAP in BC invasion, which will be highly significant for the design/synthesis of new BIR2/BIR3-based compounds for invasive BC treatment.
Collapse
Affiliation(s)
- Jiheng Xu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Xiaohui Hua
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Rui Yang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Junlan Zhu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Zhongxian Tian
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Maowen Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Guosong Jiang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, China.
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA.
| |
Collapse
|
60
|
Zhu H, Li Y, Liu Y, Han B. Bivalent SMAC Mimetics for Treating Cancer by Antagonizing Inhibitor of Apoptosis Proteins. ChemMedChem 2019; 14:1951-1962. [DOI: 10.1002/cmdc.201900410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/10/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Hongping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine 1166 Liutai Avenue Chengdu 611137 China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Yi Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Yue Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of AntibioticsChengdu University 168 Huaguan Road Chengdu 610052 China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of PharmacyChengdu University of Traditional Chinese Medicine 1166 Liutai Avenue Chengdu 611137 China
| |
Collapse
|
61
|
Ding R, Wang X, Chen W, Li Z, Wei AL, Wang QB, Nie AH, Wang LL. WX20120108, a novel IAP antagonist, induces tumor cell autophagy via activating ROS-FOXO pathway. Acta Pharmacol Sin 2019; 40:1466-1479. [PMID: 31316176 PMCID: PMC6889436 DOI: 10.1038/s41401-019-0253-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 05/19/2019] [Indexed: 12/14/2022]
Abstract
Recently, inhibitor of apoptosis proteins (IAPs) and some IAP antagonists were found to regulate autophagy, but the underlying mechanisms remain unclear. WX20120108 is an analogue of GDC-0152 (a known IAP antagonist) and displays more potent anti-tumor and autophagy-regulating activity in tumor cells, we investigated the regulatory mechanisms underlying WX20120108-induced autophagy. Using molecular docking and fluorescence polarization anisotropy (FPA) competitive assay, we first demonstrated that WX20120108, acting as an IAP antagonist, bound to the XIAP-BIR3, XIAP BIR2-BIR3, cIAP1 BIR3, and cIAP2 BIR3 domains with high affinities. In six cancer cell lines, WX20120108 inhibited the cell proliferation with potencies two to ten-fold higher than that of GDC-0152. In HeLa and MDA-MB-231 cells, WX20120108 induced caspase-dependent apoptosis and activated TNFα-dependent extrinsic apoptosis. On the other hand, WX20120108 induced autophagy in HeLa and MDA-MB-231 cells in dose- and time-dependent manners. We revealed that WX20120108 selectively activated Foxo3, evidenced by Foxo3 nuclear translocation in both gene modified cell line and HeLa cells, as well as the upregulated expression of Foxo3-targeted genes (Bnip3, Pik3c3, Atg5, and Atg4b), which played a key role in autophagy initiation. WX20120108-induced autophagy was significantly suppressed when Foxo3 gene was silenced. WX20120108 dose-dependently increased the generation of reactive oxygen species (ROS) in HeLa cells, and WX20120108-induced Foxo3 activation was completely blocked in the presence of catalase, a known ROS scavenger. However, WX20120108-induced ROS generation was not affected by cIAP1/2 or XIAP gene silencing. In conclusion, WX20120108-induced autophagy relies on activating ROS-Foxo3 pathway, which is independent of IAPs. This finding provides a new insight into the mechanism of IAP antagonist-mediated regulation of autophagy.
Collapse
Affiliation(s)
- Rui Ding
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - Xin Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Wei Chen
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Zhi Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Ai-Li Wei
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Qing-Bin Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Ai-Hua Nie
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Li-Li Wang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
| |
Collapse
|
62
|
Del Re DP, Amgalan D, Linkermann A, Liu Q, Kitsis RN. Fundamental Mechanisms of Regulated Cell Death and Implications for Heart Disease. Physiol Rev 2019; 99:1765-1817. [PMID: 31364924 DOI: 10.1152/physrev.00022.2018] [Citation(s) in RCA: 661] [Impact Index Per Article: 110.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.
Collapse
Affiliation(s)
- Dominic P Del Re
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Dulguun Amgalan
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Andreas Linkermann
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Qinghang Liu
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
63
|
XIAP as a Target of New Small Organic Natural Molecules Inducing Human Cancer Cell Death. Cancers (Basel) 2019; 11:cancers11091336. [PMID: 31505859 PMCID: PMC6770071 DOI: 10.3390/cancers11091336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022] Open
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is an emerging crucial therapeutic target in cancer. We report on the discovery and characterisation of small organic molecules from Piper genus plants exhibiting XIAP antagonism, namely erioquinol, a quinol substituted in the 4-position with an alkenyl group and the alkenylphenols eriopodols A–C. Another isolated compound was originally identified as gibbilimbol B. Erioquinol was the most potent inhibitor of human cancer cell viability when compared with gibbilimbol B and eriopodol A was listed as intermediate. Gibbilimbol B and eriopodol A induced apoptosis through mitochondrial permeabilisation and caspase activation while erioquinol acted on cell fate via caspase-independent/non-apoptotic mechanisms, likely involving mitochondrial dysfunctions and aberrant generation of reactive oxygen species. In silico modelling and molecular approaches suggested that all molecules inhibit XIAP by binding to XIAP-baculoviral IAP repeat domain. This demonstrates a novel aspect of XIAP as a key determinant of tumour control, at the molecular crossroad of caspase-dependent/independent cell death pathway and indicates molecular aspects to develop tumour-effective XIAP antagonists.
Collapse
|
64
|
Polykretis P, Luchinat E, Bonucci A, Giachetti A, Graewert MA, Svergun DI, Banci L. Conformational characterization of full-length X-chromosome-linked inhibitor of apoptosis protein (XIAP) through an integrated approach. IUCRJ 2019; 6:948-957. [PMID: 31576227 PMCID: PMC6760453 DOI: 10.1107/s205225251901073x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
The X-chromosome-linked inhibitor of apoptosis protein (XIAP) is a multidomain protein whose main function is to block apoptosis by caspase inhibition. XIAP is also involved in other signalling pathways, including NF-κB activation and copper homeostasis. XIAP is overexpressed in tumours, potentiating cell survival and resistance to chemotherapeutics, and has therefore become an important target for the treatment of malignancy. Despite the fact that the structure of each single domain is known, the conformation of the full-length protein has never been determined. Here, the first structural model of the full-length XIAP dimer, determined by an integrated approach using nuclear magnetic resonance, small-angle X-ray scattering and electron paramagnetic resonance data, is presented. It is shown that XIAP adopts a compact and relatively rigid conformation, implying that the spatial arrangement of its domains must be taken into account when studying the interactions with its physiological partners and in developing effective inhibitors.
Collapse
Affiliation(s)
- Panagis Polykretis
- CERM – Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Enrico Luchinat
- CERM – Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Alessio Bonucci
- CERM – Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Andrea Giachetti
- CERM – Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Melissa A. Graewert
- EMBL, Hamburg Outstation, European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - Dmitri I. Svergun
- EMBL, Hamburg Outstation, European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - Lucia Banci
- CERM – Magnetic Resonance Center, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
65
|
Micewicz ED, Nguyen C, Micewicz A, Waring AJ, McBride WH, Ruchala P. Position of lipidation influences anticancer activity of Smac analogs. Bioorg Med Chem Lett 2019; 29:1628-1635. [PMID: 31047753 PMCID: PMC6625762 DOI: 10.1016/j.bmcl.2019.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
A small group of lipid-conjugated Smac mimetics was synthesized to probe the influence of the position of lipidation on overall anti-cancer activity. Specifically, new compounds were modified with lipid(s) in position 3 and C-terminus. Previously described position 2 lipidated analog M11 was also synthesized. The resulting mini library of Smacs lipidated in positions 2, 3 and C-terminus was screened extensively in vitro against a total number of 50 diverse cancer cell lines revealing that both the position of lipidation as well as the type of lipid, influence their anti-cancer activity and cancer type specificity. Moreover, when used in combination therapy with inhibitor of menin-MLL1 protein interactions, position 2 modified analog SM2 showed strong synergistic anti-cancer properties. The most promising lipid-conjugated analogs SM2 and SM6, showed favorable pharmacokinetics and in vivo activity while administered subcutaneously in the preclinical mouse model. Collectively, our findings suggest that lipid modification of Smacs may be a viable approach in the development of anti-cancer therapeutic leads.
Collapse
Affiliation(s)
- Ewa D Micewicz
- Department of Radiation Oncology, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Christine Nguyen
- Department of Radiation Oncology, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Alina Micewicz
- David Geffen School of Medicine at UCLA, Volunteering Program, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Alan J Waring
- Department of Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90502, USA
| | - William H McBride
- Department of Radiation Oncology, University of California at Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Piotr Ruchala
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, 760 Westwood Plaza, Los Angeles, CA 90024, USA; The Pasarow Mass Spectrometry Laboratory, The Jane and Terry Semel Institute for Neuroscience and Human Behavior, 760 Westwood Plaza, Los Angeles, CA 90024, USA.
| |
Collapse
|
66
|
Chen SN, Fang T, Kong JY, Pan BB, Su XC. Third BIR domain of XIAP binds to both Cu(II) and Cu(I) in multiple sites and with diverse affinities characterized at atomic resolution. Sci Rep 2019; 9:7428. [PMID: 31092843 PMCID: PMC6520397 DOI: 10.1038/s41598-019-42875-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/08/2019] [Indexed: 12/05/2022] Open
Abstract
The X-chromosome linked inhibitor of apoptosis, XIAP, is mainly known as the inhibitor of caspases by direct interaction with caspases with its baculoviral IAP repeat (BIR) domains. XIAP has three BIR domains and each BIR domain contains a zinc binding site, normally known as zinc finger motif. Recent studies showed that XIAP is involved in copper homeostasis in cells and the BIR domains bind copper ion. However, structural details of the second and third BIR domain, BIR2 and BIR3, in XIAP, with copper as well as the binding modes are not known. In the present work we characterize the structural properties of BIR3 in solution by high resolution NMR and other biophysical techniques. The interaction of BIR3 with copper both in vitro and in cell lysates was analyzed. Our results show that BIR3 is able to form stable complexes both with Cu(II) and Cu(I), whereas zinc binding site is not affected and zinc retains tightly bound in the zinc finger during these interactions. Surprisingly, BIR3 has multiple binding sites for Cu(II) and Cu(I) but with varied binding affinities. In addition, the solvent exposed Cys351 is readily oxidized by Cu(II) resulting an intermolecular disulfide bond either between two BIR3 molecules or a mixed disulfide bond with glutathione in cell lysates.
Collapse
Affiliation(s)
- Shen-Na Chen
- State Key Laboratory of Elemento-Organic Chemistry, Department of Chemical Biology, College of Chemistry and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Tian Fang
- State Key Laboratory of Elemento-Organic Chemistry, Department of Chemical Biology, College of Chemistry and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Jing-Yang Kong
- State Key Laboratory of Elemento-Organic Chemistry, Department of Chemical Biology, College of Chemistry and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Bin-Bin Pan
- State Key Laboratory of Elemento-Organic Chemistry, Department of Chemical Biology, College of Chemistry and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Xun-Cheng Su
- State Key Laboratory of Elemento-Organic Chemistry, Department of Chemical Biology, College of Chemistry and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China.
| |
Collapse
|
67
|
Huang Q, Peng Y, Peng Y, Wei D, Wei Y, Feng S. The TwistDock workflow for evaluation of bivalent Smac mimetics targeting XIAP. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1373-1388. [PMID: 31118573 PMCID: PMC6499140 DOI: 10.2147/dddt.s194276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/05/2019] [Indexed: 01/13/2023]
Abstract
Purpose: Mimetics based on Smac, the native inhibitor of XIAP, are promising drug-candidates for the treatment of cancer. Bivalent Smac mimetics inhibit XIAP with even higher potency than monovalent mimetics, but how to optimize the linker that tethers the two monovalent binding motifs remains controversial. Methods: To construct an ensemble of bivalent complex structures for evaluating various linkers, we propose herein a workflow, named TwistDock, consisting of steps of monovalent docking and linker twisting, in which the degrees of freedom are sampled focusing on the rotation of single bonds of the linker. Results: The obtained conformations of bivalent complex distribute randomly in the conformational space with respect to two reaction coordinates introduced by the linker, which are the distance of the two binding motifs and the dihedral angle of the two planes through the linker and each of the binding motifs. Molecular dynamics starting from 10 conformations with the lowest enthalpy of every complex shows that the conformational tendency of the complex participated by compound 9, one of the compounds with the largest binding affinity, is distinct from others. By umbrella sampling of the complex, we find its global minimum of the free energy landscape. The structure shows that the linker favors a compact conformation, and the two BIR domains of XIAP encompass the ligand on the opposite sides. Conclusion: TwistDock can be used in fine-tuning of bivalent ligands targeting XIAP and similar receptors dimerized or oligomerized.
Collapse
Affiliation(s)
- Qingsheng Huang
- Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology and Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, People's Republic of China
| | - Yin Peng
- Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yuefeng Peng
- Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology and Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, People's Republic of China.,Center for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, MD 20903, USA
| | - Dan Wei
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Yanjie Wei
- Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology and Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, People's Republic of China
| | - Shengzhong Feng
- Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology and Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
68
|
Menacing Mold: Recent Advances in Aspergillus Pathogenesis and Host Defense. J Mol Biol 2019; 431:4229-4246. [PMID: 30954573 DOI: 10.1016/j.jmb.2019.03.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/21/2019] [Accepted: 03/30/2019] [Indexed: 02/08/2023]
Abstract
The genus Aspergillus is ubiquitous in the environment and contains a number of species, primarily A. fumigatus, that cause mold-associated disease in humans. Humans inhale several hundred to several thousand Aspergillus conidia (i.e., vegetative spores) daily and typically clear these in an asymptomatic manner. In immunocompromised individuals, Aspergillus conidia can germinate into tissue-invasive hyphae, disseminate, and cause invasive aspergillosis. In this review, we first discuss novel concepts in host defense against Aspergillus infections and emphasize new insights in fungal recognition and signaling, innate immune activation, and fungal killing. Second, the review focuses on novel concepts of Aspergillus pathogenesis and highlights emerging knowledge regarding fungal strain heterogeneity, stress responses, and metabolic adaptations on infectious outcomes. Mechanistic insight into the host-pathogen interplay is thus critical to define novel druggable fungal targets and to exploit novel immune-based strategies to improve clinical outcomes associated with aspergillosis in vulnerable patient populations.
Collapse
|
69
|
Clinical candidates modulating protein-protein interactions: The fragment-based experience. Eur J Med Chem 2019; 167:76-95. [DOI: 10.1016/j.ejmech.2019.01.084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/23/2022]
|
70
|
Shu K, Iwamoto N, Honda K, Kondoh Y, Hirano H, Osada H, Ohno H, Fujii N, Oishi S. Development of Mirror-Image Screening Systems for XIAP BIR3 Domain Inhibitors. Bioconjug Chem 2019; 30:1395-1404. [DOI: 10.1021/acs.bioconjchem.9b00154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Keitou Shu
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Graduate School of Advanced Integrated Studies in Human Survivability, Kyoto University, Sakyo-ku, Kyoto 606-8306, Japan
| | - Naoya Iwamoto
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kaori Honda
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Yasumitsu Kondoh
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Hirano
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Nobutaka Fujii
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
71
|
Gao X, Zhang L, Wei Y, Yang Y, Li J, Wu H, Yin Y. Prognostic Value of XIAP Level in Patients with Various Cancers: A Systematic Review and Meta-Analysis. J Cancer 2019; 10:1528-1537. [PMID: 31031863 PMCID: PMC6485232 DOI: 10.7150/jca.28229] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023] Open
Abstract
Background: X-linked inhibitor of apoptosis protein (XIAP) plays an important role in cancer pathogenesis, which has been found to be overexpressed in multiple human cancers and associated with survival rates. Herein, we performed a meta-analysis to explore the predictive value of XIAP level in patients with various solid tumors. Methods: Relevant articles exploring the relationship between XIAP expression and survival of cancer patients were retrieved in PubMed, PMC, EMBASE and Web of Science published from 2001 to 2018. The combined hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to evaluate the significance. Results: A total of 6554 patients from 40 articles were included in this meta-analysis. It was shown in 37 studies with 4864 cases that the over-expression of XIAP was associated with poorer overall survival (OS) (combined HR=1.61, 95% CI: 1.33-1.96). Meanwhile, 8 studies with 1862 cases revealed that elevated XIAP level predicted shorter disease-free survival (DFS) (HR=2.17, 95% CI: 1.03-4.59). Subgroup analyses showed that higher XIAP detection was related to worse OS in gastric cancer (HR=1.42, 95% CI: 1.18-1.72) and head and neck cancer (HNC) (HR=2.97, 95% CI: 1.97-4.47). Conclusion: Our results suggested that elevated XIAP level seemed to represent an unfavorable prognostic factor for clinical outcomes in cancer patients. However, there were limited studies describing the association between XIAP expression and clinical prognosis in each different type of tumors. Therefore, concrete roles of XIAP in various cancers need to be further explored.
Collapse
Affiliation(s)
- Xian Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yong Wei
- Department of Urology, Nanjing Gaochun People's Hospital, Nanjing, 211300, China
| | - Yiqi Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
72
|
Targeting the BIR Domains of Inhibitor of Apoptosis (IAP) Proteins in Cancer Treatment. Comput Struct Biotechnol J 2019; 17:142-150. [PMID: 30766663 PMCID: PMC6360406 DOI: 10.1016/j.csbj.2019.01.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 01/07/2023] Open
Abstract
Inhibitor of apoptosis (IAP) proteins are characterized by the presence of the conserved baculoviral IAP repeat (BIR) domain that is involved in protein-protein interactions. IAPs were initially thought to be mainly responsible for caspase inhibition, acting as negative regulators of apoptosis, but later works have shown that IAPs also control a plethora of other different cellular pathways. As X-linked IAP (XIAP), and other IAP, levels are often deregulated in cancer cells and have been shown to correlate with patients' prognosis, several approaches have been pursued to inhibit their activity in order to restore apoptosis. Many small molecules have been designed to target the BIR domains, the vast majority being inspired by the N-terminal tetrapeptide of Second Mitochondria-derived Activator of Caspases/Direct IAp Binding with Low pI (Smac/Diablo), which is the natural XIAP antagonist. These compounds are therefore usually referred to as Smac mimetics (SMs). Despite the fact that SMs were intended to specifically target XIAP, it has been shown that they also interact with cellular IAP-1 (cIAP1) and cIAP2, promoting their proteasome-dependent degradation. SMs have been tested in combination with several cytotoxic compounds and are now considered promising immune modulators which can be exploited in cancer therapy, especially in combination with immune checkpoint inhibitors. In this review, we give an overview of the structural hot-spots of BIRs, focusing on their fold and on the peculiar structural patches which characterize the diverse BIRs. These structures are exploited/exploitable for the development of specific and active IAP inhibitors.
Collapse
|
73
|
Sheng ZJ, Shi YM, Xu X, Bellynck S, Zhang K, Du ZY, Xu X, Maurel F, Dong CZ. Development of XIAP Antagonists Based On De Novo 8,5-Fused Bicyclic Lactams. ChemistryOpen 2019; 8:34-40. [PMID: 30652062 PMCID: PMC6331714 DOI: 10.1002/open.201800260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
In order to develop original water soluble antagonists of X‐linked inhibitor of apoptosis protein (XIAP), a novel bicyclic scaffold was designed based on 8,5‐fused bicyclic lactam. During its preparation, a spontaneous rearrangement from 8,5‐ to 7,5‐fused bicyclic lactam was observed and confirmed by MS and NMR analyses, in particular the HMBC spectra. DFT calculations were performed to understand the corresponding mechanism. It was finally prevented through changing the reaction order in the synthesis route and a Smac mimetic with this core structure, ZJ‐1 was successfully obtained. The structure of this new bicyclic scaffold was well confirmed by HRMS and NMR (1H, 13C, NOESY) analyses. ZJ‐1 presented in addition a binding affinity to XIAP‐BIR3, nearly 6 times better than that of AVPI, similar to the reported SM‐128 in an in vitro fluorescence polarization (FP) assay. This preliminary result suggests that this new bicyclic scaffold could be very attractive in the development of novel anticancer agents targeting XIAP.
Collapse
Affiliation(s)
- Zhaojun J Sheng
- Interfaces, Traitements, Organisation et Dynamique des Systemes (ITODYS) UMR 7086 CNRS Université Paris Diderot, Sorbonne Paris Cité 75205 Paris France.,School of Chemical Engineering and Light Industry Institute of Natural Medicine and Green Chemistry Guangdong University of Technology 510006 Guangzhou China.,School of Chemical and Environment Engineering Wuyi University 529020 Jiangmen China
| | - Yiming M Shi
- Interfaces, Traitements, Organisation et Dynamique des Systemes (ITODYS) UMR 7086 CNRS Université Paris Diderot, Sorbonne Paris Cité 75205 Paris France
| | - Ximing Xu
- School of Electrical and Information Engineering Institute of Bioinformatics and Medical Engineering Jiangsu University of Technology 213001 Changzhou China
| | - Sébastien Bellynck
- Interfaces, Traitements, Organisation et Dynamique des Systemes (ITODYS) UMR 7086 CNRS Université Paris Diderot, Sorbonne Paris Cité 75205 Paris France
| | - Kun Zhang
- School of Chemical Engineering and Light Industry Institute of Natural Medicine and Green Chemistry Guangdong University of Technology 510006 Guangzhou China.,School of Chemical and Environment Engineering Wuyi University 529020 Jiangmen China
| | - Zhiyun Y Du
- School of Chemical Engineering and Light Industry Institute of Natural Medicine and Green Chemistry Guangdong University of Technology 510006 Guangzhou China
| | - Xuetao Xu
- School of Chemical and Environment Engineering Wuyi University 529020 Jiangmen China
| | - François Maurel
- Interfaces, Traitements, Organisation et Dynamique des Systemes (ITODYS) UMR 7086 CNRS Université Paris Diderot, Sorbonne Paris Cité 75205 Paris France
| | - Chang-Zhi Dong
- Interfaces, Traitements, Organisation et Dynamique des Systemes (ITODYS) UMR 7086 CNRS Université Paris Diderot, Sorbonne Paris Cité 75205 Paris France.,School of Chemical Engineering and Light Industry Institute of Natural Medicine and Green Chemistry Guangdong University of Technology 510006 Guangzhou China.,Institute for Interdisciplinary Research Jianghan University 430056 Wuhan China
| |
Collapse
|
74
|
Tang HM, Tang HL. Anastasis: recovery from the brink of cell death. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180442. [PMID: 30839720 PMCID: PMC6170572 DOI: 10.1098/rsos.180442] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/23/2018] [Indexed: 05/11/2023]
Abstract
Anastasis is a natural cell recovery phenomenon that rescues cells from the brink of death. Programmed cell death such as apoptosis has been traditionally assumed to be an intrinsically irreversible cascade that commits cells to a rapid and massive demolition. Interestingly, recent studies have demonstrated recovery of dying cells even at the late stages generally considered immutable. Here, we examine the evidence for anastasis in cultured cells and in animals, review findings illuminating the potential mechanisms of action, discuss the challenges of studying anastasis and explore new strategies to uncover the function and regulation of anastasis, the identification of which has wide-ranging physiological, pathological and therapeutic implications.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
75
|
Martos-Maldonado MC, Hjuler CT, Sørensen KK, Thygesen MB, Rasmussen JE, Villadsen K, Midtgaard SR, Kol S, Schoffelen S, Jensen KJ. Selective N-terminal acylation of peptides and proteins with a Gly-His tag sequence. Nat Commun 2018; 9:3307. [PMID: 30120230 PMCID: PMC6098153 DOI: 10.1038/s41467-018-05695-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
Methods for site-selective chemistry on proteins are in high demand for the synthesis of chemically modified biopharmaceuticals, as well as for applications in chemical biology, biosensors and more. Inadvertent N-terminal gluconoylation has been reported during expression of proteins with an N-terminal His tag. Here we report the development of this side-reaction into a general method for highly selective N-terminal acylation of proteins to introduce functional groups. We identify an optimized N-terminal sequence, GHHHn- for the reaction with gluconolactone and 4-methoxyphenyl esters as acylating agents, facilitating the introduction of functionalities in a highly selective and efficient manner. Azides, biotin or a fluorophore are introduced at the N-termini of four unrelated proteins by effective and selective acylation with the 4-methoxyphenyl esters. This Gly-Hisn tag adds the unique capability for highly selective N-terminal chemical acylation of expressed proteins. We anticipate that it can find wide application in chemical biology and for biopharmaceuticals.
Collapse
Affiliation(s)
- Manuel C Martos-Maldonado
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Christian T Hjuler
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Kasper K Sørensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Mikkel B Thygesen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Jakob E Rasmussen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Klaus Villadsen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark
| | - Søren R Midtgaard
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Stefan Kol
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet Building 220, 2800, Kgs. Lyngby, Denmark
| | - Sanne Schoffelen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark. .,Center for Evolutionary Chemical Biology, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark.
| | - Knud J Jensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark. .,Biomolecular Nanoscale Engineering Center, University of Copenhagen, Thorvaldsensvej 40, 1871, Frederiksberg, Denmark.
| |
Collapse
|
76
|
Chirieleison SM, Rathkey JK, Abbott DW. Unique BIR domain sets determine inhibitor of apoptosis protein-driven cell death and NOD2 complex signal specificity. Sci Signal 2018; 11:11/539/eaao3964. [PMID: 30018081 DOI: 10.1126/scisignal.aao3964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mammalian IAPs, X-linked inhibitor of apoptosis protein (XIAP) and cellular inhibitor of apoptosis protein 1 and 2 (cIAP1 and cIAP2), play pivotal roles in innate immune signaling and inflammatory homeostasis, often working in parallel or in conjunction at a signaling complex. IAPs direct both nucleotide-binding oligomerization domain-containing 2 (NOD2) signaling complexes and cell death mechanisms to appropriately regulate inflammation. Although it is known that XIAP is critical for NOD2 signaling and that the loss of cIAP1 and cIAP2 blunts NOD2 activity, it is unclear whether these three highly related proteins can compensate for one another in NOD2 signaling or in mechanisms governing apoptosis or necroptosis. This potential redundancy is critically important, given that genetic loss of XIAP causes both very early onset inflammatory bowel disease and X-linked lymphoproliferative syndrome 2 (XLP-2) and that the overexpression of cIAP1 and cIAP2 is linked to both carcinogenesis and chemotherapeutic resistance. Given the therapeutic interest in IAP inhibition and the potential toxicities associated with disruption of inflammatory homeostasis, we used synthetic biology techniques to examine the functional redundancies of key domains in the IAPs. From this analysis, we defined the features of the IAPs that enable them to function at overlapping signaling complexes but remain independent and functionally exclusive in their roles as E3 ubiquitin ligases in innate immune and inflammatory signaling.
Collapse
Affiliation(s)
- Steven M Chirieleison
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44122, USA
| | - Joseph K Rathkey
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44122, USA
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44122, USA.
| |
Collapse
|
77
|
Aouacheria A, Cunningham KW, Hardwick JM, Palková Z, Powers T, Severin FF, Váchová L. Comment on "Sterilizing immunity in the lung relies on targeting fungal apoptosis-like programmed cell death". Science 2018; 360:360/6395/eaar6910. [PMID: 29930109 DOI: 10.1126/science.aar6910] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/10/2018] [Indexed: 12/22/2022]
Abstract
Shlezinger et al (Reports, 8 September 2017, p. 1037) report that the common fungus Aspergillus fumigatus, a cause of aspergillosis, undergoes caspase-dependent apoptosis-like cell death triggered by lung neutrophils. However, the technologies they used do not provide reliable evidence that fungal cells die via a protease signaling cascade thwarted by a fungal caspase inhibitor homologous to human survivin.
Collapse
Affiliation(s)
- Abdel Aouacheria
- ISEM, Institut des Sciences de l'Evolution de Montpellier, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Kyle W Cunningham
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD 21205, USA.
| | - Zdena Palková
- Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ted Powers
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Fedor F Severin
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119991, Russia
| | - Libuše Váchová
- Institute of Microbiology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
78
|
Over-activation of AKT signaling leading to 5-Fluorouracil resistance in SNU-C5/5-FU cells. Oncotarget 2018; 9:19911-19928. [PMID: 29731993 PMCID: PMC5929436 DOI: 10.18632/oncotarget.24952] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/13/2018] [Indexed: 12/13/2022] Open
Abstract
Here, we investigated whether over-activation of AKT pathway is important in the resistance to 5-fluorouracil (5-FU) in SNU-C5/5-FU cells, 5-FU-resistant human colon cancer cells. When compared to wild type SNU-C5 cells (WT), SNU-C5/5-FU cells showed over-activation of PI3K/AKT pathway, like increased phosphorylation of AKT, mTOR, and GSK-3β, nuclear localization of β-catenin, and decreased E-cadherin. Moreover, E-cadherin level was down-regulated in recurrent colon cancer tissues compared to primary colon cancer tissues. Gene silencing of AKT1 or treatment of LY294002 (PI3 kinase inhibitor) increased E-cadherin, whereas decreased phospho-GSK-3β. LY294002 also reduced protein level of β-catenin with no influence on mRNA level. PTEN level was higher in SNU-C5/WT than SNU-C5/5-FU cells, whereas the loss of PETN in SNU-C5/WT cells induced characteristics of SNU-C5/5-FU cells. In SNU-C5/5-FU cells, NF-κB signaling was activated, along with the overexpression of COX-2 and stabilization of survivin. However, increased COX-2 contributed to the stabilization of survivin, which directly interacts with cytoplasmic procaspase-3, while the inhibition of AKT reduced this cascade. We finally confirmed that combination treatment with 5-FU and LY294002 or Vioxx could induce apoptosis in SNU-C5/5-FU cells. These data suggest that inhibition of AKT activation may overcome 5-FU-resistance in SNU-C5/5-FU cells. These findings provide evidence that over-activation of AKT is crucial for the acquisition of resistance to anticancer drugs and AKT pathway could be a therapeutic target for cancer treatment.
Collapse
|
79
|
The Autism Protein Ube3A/E6AP Remodels Neuronal Dendritic Arborization via Caspase-Dependent Microtubule Destabilization. J Neurosci 2017; 38:363-378. [PMID: 29175955 DOI: 10.1523/jneurosci.1511-17.2017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/27/2017] [Accepted: 10/31/2017] [Indexed: 02/01/2023] Open
Abstract
UBE3A gene copy number variation and the resulting overexpression of the protein E6AP is directly linked to autism spectrum disorders (ASDs). However, the underlying cellular and molecular neurobiology remains less clear. Here we report the role of ASD-related increased dosage of Ube3A/E6AP in dendritic arborization during brain development. We show that increased E6AP expression in primary cultured neurons leads to a reduction in dendritic branch number and length. The E6AP-dependent remodeling of dendritic arborization results from retraction of dendrites by thinning and fragmentation at the tips of dendrite branches, leading to shortening or removal of dendrites. This remodeling effect is mediated by the ubiquitination and degradation of XIAP (X-linked inhibitors of aptosis protein) by E6AP, which leads to activation of caspase-3 and cleavage of microtubules. In vivo, male and female Ube3A 2X ASD mice show decreased XIAP levels, increased caspase-3 activation, and elevated levels of tubulin cleavage. Consistently, dendritic branching and spine density are reduced in cortical neurons of Ube3A 2X ASD mice. In revealing an important role for Ube3A/E6AP in ASD-related developmental alteration in dendritic arborization and synapse formation, our findings provide new insights into the pathogenesis of Ube3A/E6AP-dependent ASD.SIGNIFICANCE STATEMENT Copy number variation of the UBE3A gene and aberrant overexpression of the gene product E6AP protein is a common cause of autism spectrum disorders (ASDs). During brain development, dendritic growth and remodeling play crucial roles in neuronal connectivity and information integration. We found that in primary neurons and in Ube3A transgenic autism mouse brain, overexpression of E6AP leads to significant loss of dendritic arborization. This effect is mediated by the ubiquitination of XIAP (X-linked inhibitor of aptosis protein) by E6AP, subsequent activation of caspases, and the eventual cleavage of microtubules, leading to local degeneration and retraction at the tips of dendritic branches. These findings demonstrate dysregulation in neuronal structural stability as a major cellular neuropathology in ASD.
Collapse
|
80
|
Hou MM, Polykretis P, Luchinat E, Wang X, Chen SN, Zuo HH, Yang Y, Chen JL, Ye Y, Li C, Banci L, Su XC. Solution structure and interaction with copper in vitro and in living cells of the first BIR domain of XIAP. Sci Rep 2017; 7:16630. [PMID: 29192194 PMCID: PMC5709467 DOI: 10.1038/s41598-017-16723-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022] Open
Abstract
The X-chromosome linked inhibitor of apoptosis (XIAP) is a multidomain metalloprotein involved in caspase inhibition and in copper homeostasis. It contains three zinc-binding baculoviral IAP repeats (BIR) domains, which are responsible for caspase interaction. Recently, it has been suggested that the BIR domains can bind copper, however high resolution data on such interaction is missing. Here we characterize by NMR the structural properties of BIR1 in solution, and the effects of its interaction with copper both in vitro and in physiological environments. BIR1 is dimeric in solution, consistent with the X-ray structure. Cysteine 12, located in the unfolded N-terminal region, has a remarkably low redox potential, and is prone to oxidation even in reducing physiological environments. Interaction of BIR1 with copper(II) results in the oxidation of cysteine 12, with the formation of either an intermolecular disulfide bond between two BIR1 molecules or a mixed disulfide bond with glutathione, whereas the zinc binding site is not affected by the interaction.
Collapse
Affiliation(s)
- Meng-Meng Hou
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Panagis Polykretis
- Magnetic Resonance Center - CERM, University of Florence, 50019, Sesto Fiorentino, Florence, Italy
| | - Enrico Luchinat
- Magnetic Resonance Center - CERM, University of Florence, 50019, Sesto Fiorentino, Florence, Italy
- Department of Biomedical, Clinical and Experimental Sciences, University of Florence, 50134, Florence, Italy
| | - Xiao Wang
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Shen-Na Chen
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Hui-Hui Zuo
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Yin Yang
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Jia-Liang Chen
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China
| | - Yansheng Ye
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Conggang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lucia Banci
- Magnetic Resonance Center - CERM, University of Florence, 50019, Sesto Fiorentino, Florence, Italy.
- Department of Chemistry, University of Florence, 50019, Sesto Fiorentino, Florence, Italy.
| | - Xun-Cheng Su
- State Key Laboratory and Research Institute of Elemento-organic Chemistry, College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, China.
| |
Collapse
|
81
|
Ono H, Iizumi Y, Goi W, Sowa Y, Taguchi T, Sakai T. Ribosomal protein S3 regulates XIAP expression independently of the NF-κB pathway in breast cancer cells. Oncol Rep 2017; 38:3205-3210. [DOI: 10.3892/or.2017.6008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/15/2017] [Indexed: 11/06/2022] Open
|
82
|
Kim SY, Park S, Yoo S, Rho JK, Jun ES, Chang S, Kim KK, Kim SC, Kim I. Downregulation of X-linked inhibitor of apoptosis protein by '7-Benzylidenenaltrexone maleate' sensitizes pancreatic cancer cells to TRAIL-induced apoptosis. Oncotarget 2017; 8:61057-61071. [PMID: 28977846 PMCID: PMC5617406 DOI: 10.18632/oncotarget.17841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/14/2017] [Indexed: 12/19/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potential biological anticancer agent. However, a wide range of human primary cancers, including pancreatic cancer, display resistance to apoptosis induction by TRAIL. Therefore, this resistance needs to be overcome to allow TRAIL to be successfully used in cancer therapy. In this study, we performed a compound screen to isolate TRAIL sensitizers and found that one of the identified compounds, 7-benzylidenenaltrexone maleate (BNTX), sensitized pancreatic cancer cells to TRAIL-induced apoptotic cell death. The combination of BNTX with TRAIL promoted the release of cytochrome c from mitochondria into cytosol with caspase activation and a resulting increase in annexin V-stained cells. From a mechanistic perspective, we found that BNTX downregulated X-linked inhibitor of apoptosis protein (XIAP) expression when used in combination with TRAIL, and found that TRAIL-induced apoptosis was augmented by siRNA-mediated knockdown of XIAP. We further demonstrated that BNTX promoted the ubiquitin/proteasome-dependent degradation of XIAP protein via protein kinase C (PKC) alpha/AKT pathway inhibition. Moreover, combined treatment by BNTX with TRAIL suppressed growth of pancreatic tumor xenograft of animal model. Therefore, we suggest that inhibitor of apoptosis protein-mediated resistance of pancreatic cancer cells to anticancer therapeutics can be overcome by inhibiting the PKCα/AKT pathway.
Collapse
Affiliation(s)
- So Young Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
| | - Sojung Park
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
| | - SeonA Yoo
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
| | - Jin Kyung Rho
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun Sung Jun
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Suhwan Chang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Kyung Kon Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Song Cheol Kim
- Division of HBP Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, South Korea
| | - Inki Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505 South Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| |
Collapse
|
83
|
Martini C, Bédard M, Lavigne P, Denault JB. Characterization of Hsp90 Co-Chaperone p23 Cleavage by Caspase-7 Uncovers a Peptidase–Substrate Interaction Involving Intrinsically Disordered Regions. Biochemistry 2017; 56:5099-5111. [DOI: 10.1021/acs.biochem.7b00298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Cyrielle Martini
- Department
of Pharmacology-Physiology and ‡Department of Biochemistry, Institut
de Pharmacologie de Sherbrooke, Université de Sherbrooke, Faculty of Medicine and Health Sciences, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4, Canada
| | - Mikaël Bédard
- Department
of Pharmacology-Physiology and ‡Department of Biochemistry, Institut
de Pharmacologie de Sherbrooke, Université de Sherbrooke, Faculty of Medicine and Health Sciences, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4, Canada
| | - Pierre Lavigne
- Department
of Pharmacology-Physiology and ‡Department of Biochemistry, Institut
de Pharmacologie de Sherbrooke, Université de Sherbrooke, Faculty of Medicine and Health Sciences, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-Bernard Denault
- Department
of Pharmacology-Physiology and ‡Department of Biochemistry, Institut
de Pharmacologie de Sherbrooke, Université de Sherbrooke, Faculty of Medicine and Health Sciences, 3001, 12th Avenue North, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
84
|
Gu C, Wang Z, Jin Z, Li G, Kou Y, Jia Z, Yang J, Tian F. MicroRNA-212 inhibits the proliferation, migration and invasion of renal cell carcinoma by targeting X-linked inhibitor of apoptosis protein (XIAP). Oncotarget 2017; 8:92119-92133. [PMID: 29190902 PMCID: PMC5696168 DOI: 10.18632/oncotarget.20786] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/28/2017] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs have been found to be critical regulator of cancer cell biology. MicroRNA-212 (miR-212) was identified to be a critical cancer-associated microRNA playing either oncogenic functions or tumor suppressive roles in different types of human cancers. In this study, we found that the level of miR-212 in renal cell carcinoma (RCC) tissues was significantly lower than that in adjacent non-tumor tissues. Decreased level of miR-212 was associated with advanced T stage and TNM stage of RCC. The expression of miR-212 was decreased in RCC cell lines as compared with the HK-2 cell line. Overexpression of miR-212 inhibited cell viability, proliferation, migration and invasion of CAKI-2 cells. Knockdown of miR-212 increased cell viability and proliferation, migration and invasion of ACHN cells. In vivo experiments showed that miR-212 inhibited the proliferation and promoted the apoptosis of ACHN cells in nude mice and thus inhibited the in vivo tumor growth of CAKI-2 cells. Furthermore, we confirmed that X-linked inhibitor of apoptosis protein (XIAP) was the downstream target of miR-212. The expression level of miR-212 was negatively correlated with XIAP expression in RCC tissues. Moreover, XIAP mediated the tumor suppressive roles of miR-212 in RCC. Finally, we demonstrated that the aberrant expression of miR-212 and XIAP was evidently correlated with poor prognosis of RCC patients. In all, miR-212 can act as a prognostic biomarker for RCC patients and inhibits the growth and metastasis of RCC cells by inhibiting XIAP.
Collapse
Affiliation(s)
- Chaohui Gu
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhiyu Wang
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhibo Jin
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guanru Li
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yiping Kou
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhankui Jia
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jinjian Yang
- Department of Urology and Henan Institute of Urology, Zhengzhou Key Laboratory for Molecular Biology of Urological Tumor Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fengyan Tian
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
85
|
The E3 ligase HECTD3 promotes esophageal squamous cell carcinoma (ESCC) growth and cell survival through targeting and inhibiting caspase-9 activation. Cancer Lett 2017; 404:44-52. [DOI: 10.1016/j.canlet.2017.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 01/31/2023]
|
86
|
Hideshima T, Mazitschek R, Qi J, Mimura N, Tseng JC, Kung AL, Bradner JE, Anderson KC. HDAC6 inhibitor WT161 downregulates growth factor receptors in breast cancer. Oncotarget 2017; 8:80109-80123. [PMID: 29113288 PMCID: PMC5655183 DOI: 10.18632/oncotarget.19019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/08/2017] [Indexed: 02/01/2023] Open
Abstract
We have shown that WT-161, a histone deacetylase 6 (HDAC6) inhibitor, shows remarkable anti-tumor activity in multiple myeloma (MM) in preclinical models. However, its activity in other type of cancers has not yet been shown. In this study, we further evaluated the biologic sequelae of WT161 in breast cancer cell lines. WT161 triggers apoptotic cell death in MCF7, T47D, BT474, and MDA-MB231 cells, associated with decreased expression of EGFR, HER2, and ERα and downstream signaling. However, HDAC6 knockdown shows that cytotoxicity and destabilization of these receptors triggered by WT161 are not dependent on HDAC6 inhibition. Moreover WT161 analog MAZ1793, which lacks HDAC inhibitory effect, similarly triggers cell line growth inhibition and downregulation of these receptors. We also confirm that WT161 significantly inhibits in vivo MCF7 cell growth, associated with downregulation of ERα, in a murine xenograft model. Finally, WT161 synergistically enhances bortezomib-induced cytotoxicity, even in bortezomib-resistant breast cancer cells. Our results therefore provide the rationale to develop a novel class of therapeutic agents targeting growth pathways central to the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Teru Hideshima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jun Qi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Naoya Mimura
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.,Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Jen-Chieh Tseng
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA, USA.,PerkinElmer Inc., Hopkinton, MA, USA
| | - Andrew L Kung
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children's Hospital Boston, Boston, MA, USA.,Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
87
|
Alves J, Garay-Malpartida M, Occhiucci JM, Belizário JE. Modulation of procaspase-7 self-activation by PEST amino acid residues of the N-terminal prodomain and intersubunit linker. Biochem Cell Biol 2017; 95:634-643. [PMID: 28658581 DOI: 10.1139/bcb-2016-0220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Procaspase-7 zymogen polypeptide is composed of a short prodomain, a large subunit (p20), and a small subunit (p10) connected to an intersubunit linker. Caspase-7 is activated by an initiator caspase-8 and -9, or by autocatalysis after specific cleavage at IQAD198↓S located at the intersubunit linker. Previously, we identified that PEST regions made of amino acid residues Pro (P), Glu (E), Asp (D), Ser (S), Thr (T), Asn (N), and Gln (Q) are conserved flanking amino acid residues in the cleavage sites within a prodomain and intersubunit linker of all caspase family members. Here we tested the impact of alanine substitution of PEST amino acid residues on procaspase-7 proteolytic self-activation directly in Escherichia coli. The p20 and p10 subunit cleavage were significantly delayed in double caspase-7 mutants in the prodomain (N18A/P26A) and intersubunit linker (S199A/P201A), compared with the wild-type caspase-7. The S199A/P201A mutants effectively inhibited the p10 small subunit cleavage. However, the mutations did not change the kinetic parameters (kcat/KM) and optimal tetrapeptide specificity (DEVD) of the purified mutant enzymes. The results suggest a role of PEST-amino acid residues in the molecular mechanism for prodomain and intersubunit cleavage and caspase-7 self-activation.
Collapse
Affiliation(s)
- Juliano Alves
- a Department of Pharmacology, Institute of Biomedical Sciences, Avenida Lineu Prestes, 1524, São Paulo, SP, 05508-900, Brazil
| | - Miguel Garay-Malpartida
- b School of Arts, Communication and Humanity, University of São Paulo, Rua Arlindo Béttio, 1000, São Paulo, SP, 03828-000, Brazil
| | - João M Occhiucci
- a Department of Pharmacology, Institute of Biomedical Sciences, Avenida Lineu Prestes, 1524, São Paulo, SP, 05508-900, Brazil
| | - José E Belizário
- a Department of Pharmacology, Institute of Biomedical Sciences, Avenida Lineu Prestes, 1524, São Paulo, SP, 05508-900, Brazil
| |
Collapse
|
88
|
Barra CN, Macedo BM, Cadrobbi KG, Pulz LH, Huete GC, Kleeb SR, Xavier JG, Catão-Dias JL, Nishiya AT, Fukumasu H, Strefezzi RF. Apoptotic intrinsic pathway proteins predict survival in canine cutaneous mast cell tumours. Vet Comp Oncol 2017; 16:E38-E44. [DOI: 10.1111/vco.12330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/04/2017] [Accepted: 05/24/2017] [Indexed: 02/01/2023]
Affiliation(s)
- C. N. Barra
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia; Universidade de São Paulo; São Paulo Brazil
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - B. M. Macedo
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - K. G. Cadrobbi
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - L. H. Pulz
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia; Universidade de São Paulo; São Paulo Brazil
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - G. C. Huete
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - S. R. Kleeb
- Universidade Metodista de São Paulo; São Bernardo do Campo Brazil
| | | | - J. L. Catão-Dias
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia; Universidade de São Paulo; São Paulo Brazil
| | | | - H. Fukumasu
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| | - R. F. Strefezzi
- Laboratório de Oncologia Comparada e Translacional, Faculdade de Zootecnia e Engenharia de Alimentos; Universidade de São Paulo; Pirassununga Brazil
| |
Collapse
|
89
|
Xie Q, Lin Q, Li D, Chen J. Imatinib induces autophagy via upregulating XIAP in GIST882 cells. Biochem Biophys Res Commun 2017; 488:584-589. [PMID: 28528977 DOI: 10.1016/j.bbrc.2017.05.096] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms originating from the gastrointestinal tract with gain of function mutations in receptor tyrosine kinases KIT or platelet-derived growth factor receptor A (PDGFRA). The main effective treatment for GISTs is tyrosine kinase inhibitors, such as imatinib mesylate. However, GISTs respond to imatinib treatment eventually develop acquired resistance, which is a main obstacle for GISTs therapy. Therefore, it's urgent to have a better understanding of the mechanisms underlying the imatinib resistance in GISTs to develop novel therapeutic strategies. X-linked inhibitor of apoptosis (XIAP) is the most potent apoptosis inhibitor among the inhibitor of apoptosis protein (IAP) family members. Increased cellular expression of XIAP often leads to drug resistance in cancers. Here we report that XIAP is induced upon imatinb treatment in GIST882 cells, leading to imatinib-induced autophagy. Imatinib-induced autophagy was impaired in XIAP-knockout cells generated by CRISPR/Cas9 system demonstrated by the decreasing of LC3 lipidation. XIAP knockout sensitizes GIST882 cells to imatinib-induced apoptotic cell death, suggesting that XIAP protects GIST882 cells from imatinib-induced cell death by inducing autophagy. Thus, the resistance of the GIST882 cells to imatinib appears to be, in part, due to the increasing of XIAP and subsequent induction of autophagy.
Collapse
Affiliation(s)
- Qingqing Xie
- School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Qi Lin
- School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Dezhi Li
- School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Jianming Chen
- Key Laboratory of Marine Genetic Resources, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Third Institute of Oceanography, Xiamen, Fujian 361005, China.
| |
Collapse
|
90
|
XIAP Interacts with and Regulates the Activity of FAF1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1335-1348. [PMID: 28414080 DOI: 10.1016/j.bbamcr.2017.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 03/26/2017] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Cell death depends on the balance between the activities of pro- and anti-apoptotic factors. X-linked inhibitor of apoptosis protein (XIAP) plays an important role in the cytoprotective process by inhibiting the caspase cascade and regulating pro-survival signaling pathways. While searching for novel interacting partners of XIAP, we identified Fas-associated factor 1 (FAF1). Contrary to XIAP, FAF1 is a pro-apoptotic factor that also regulates several signaling pathways in which XIAP is involved. However, the functional relationship between FAF1 and XIAP is unknown. Here, we describe a new interaction between XIAP and FAF1 and describe the functional implications of their opposing roles in cell death and NF-κB signaling. Our results clearly demonstrate the interaction of XIAP with FAF1 and define the specific region of the interaction. We observed that XIAP is able to block FAF1-mediated cell death by interfering with the caspase cascade and directly interferes in NF-κB pathway inhibition by FAF1. Furthermore, we show that XIAP promotes ubiquitination of FAF1. Conversely, FAF1 does not interfere with the anti-apoptotic activity of XIAP, despite binding to the BIR domains of XIAP; however, FAF1 does attenuate XIAP-mediated NF-κB activation. Altered expression of both factors has been implicated in degenerative and cancerous processes; therefore, studying the balance between XIAP and FAF1 in these pathologies will aid in the development of novel therapies.
Collapse
|
91
|
Ge Y, Lei W, Ma Y, Wang Y, Wei B, Chen X, Ru G, He X, Mou X, Wang S. Synergistic antitumor effects of CDK inhibitor SNS‑032 and an oncolytic adenovirus co‑expressing TRAIL and Smac in pancreatic cancer. Mol Med Rep 2017; 15:3521-3528. [PMID: 28440486 PMCID: PMC5436152 DOI: 10.3892/mmr.2017.6472] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
Gene therapy using oncolytic adenoviruses is a novel approach for human cancer therapeutics. The current study aimed to investigate whether the combined use of an adenovirus expressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and second mitochondria-derived activator of caspase (Smac) upon caspase activation (ZD55-TRAIL-IETD-Smac) and cyclin-dependent kinase (CDK) inhibitor SNS-032 will synergistically reinforce their anti-pancreatic cancer activities. The experiments in vitro demonstrated that SNS-032 enhances ZD55-TRAIL-IETD-Smac-induced apoptosis and causes marked pancreatic cancer cell death. Western blot assays suggested that the SNS-032 intensified ZD55-TRAIL-IETD-Smac-induced apoptosis of pancreatic cancer cells by affecting anti-apoptotic signaling elements, including CDK-2, CDK-9, Mcl-1 and XIAP. Additionally, animal experiments further confirmed that the combination of SNS-032 and ZD55-TRAIL-IETD-Smac significantly inhibited the growth of BxPC-3 pancreatic tumor xenografts. In conclusion, the present study demonstrated that SNS-032 sensitizes human pancreatic cancer cells to ZD55-TRAIL-IETD-Smac-induced cell death in vitro and in vivo. These findings indicate that combined treatment with SNS-032 and ZD55-TRAIL-IETD-Smac could represent a rational approach for anti-pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yun Ge
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Wen Lei
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Buyun Wei
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Xiaoyi Chen
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xianglei He
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
92
|
Chirieleison SM, Marsh RA, Kumar P, Rathkey JK, Dubyak GR, Abbott DW. Nucleotide-binding oligomerization domain (NOD) signaling defects and cell death susceptibility cannot be uncoupled in X-linked inhibitor of apoptosis (XIAP)-driven inflammatory disease. J Biol Chem 2017; 292:9666-9679. [PMID: 28404814 DOI: 10.1074/jbc.m117.781500] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/03/2017] [Indexed: 12/22/2022] Open
Abstract
The X-linked inhibitor of apoptosis (XIAP) protein has been identified as a key genetic driver of two distinct inflammatory disorders, X-linked lymphoproliferative syndrome 2 (XLP-2) and very-early-onset inflammatory bowel disease (VEO-IBD). Molecularly, the role of XIAP mutations in the pathogenesis of these disorders is unclear. Recent work has consistently shown XIAP to be critical for signaling downstream of the Crohn's disease susceptibility protein nucleotide-binding oligomerization domain-containing 2 (NOD2); however, the reported effects of XLP-2 and VEO-IBD XIAP mutations on cell death have been inconsistent. In this manuscript, we describe a CRISPR-mediated genetic system for cells of the myeloid lineage in which XIAP alleles can be replaced with disease-associated XIAP variants expressed at endogenous levels to simultaneously study inflammation-related cell death and NOD2 signaling. We show that, consistent with previous studies, NOD2 signaling is critically dependent on the BIR2 domain of XIAP. We further used this system to reconcile the aforementioned inconsistent XIAP cell death data to show that XLP-2 and VEO-IBD XIAP mutations that exhibit a loss-of-function NOD2 phenotype also lower the threshold for inflammatory cell death. Last, we identified and studied three novel patient XIAP mutations and used this system to characterize NOD2 and cell death phenotypes driven by XIAP. The results of this work support the role of XIAP in mediating NOD2 signaling while reconciling the role of XLP-2 and VEO-IBD XIAP mutations in inflammatory cell death and provide a set of tools and framework to rapidly test newly discovered XIAP variants.
Collapse
Affiliation(s)
| | - Rebecca A Marsh
- the Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital, Cincinnati, Ohio 45229
| | | | | | - George R Dubyak
- Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106 and
| | | |
Collapse
|
93
|
Deletion Of XIAP reduces the severity of acute pancreatitis via regulation of cell death and nuclear factor-κB activity. Cell Death Dis 2017; 8:e2685. [PMID: 28300832 PMCID: PMC5386564 DOI: 10.1038/cddis.2017.70] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 01/10/2017] [Accepted: 01/24/2017] [Indexed: 02/05/2023]
Abstract
Severe acute pancreatitis (SAP) still remains a clinical challenge, not only for its high mortality but the uncontrolled inflammatory progression from acute pancreatitis (AP) to SAP. Cell death, including apoptosis and necrosis are critical pathology of AP, since the severity of pancreatitis correlates directly with necrosis and inversely with apoptosis Therefore, regulation of cell death from necrosis to apoptosis may have practicably therapeutic value. X-linked inhibitor of apoptosis protein (XIAP) is the best characterized member of the inhibitor of apoptosis proteins (IAP) family, but its function in AP remains unclear. In the present study, we investigated the potential role of XIAP in regulation of cell death and inflammation during acute pancreatitis. The in vivo pancreatitis model was induced by the administration of cerulein with or without lipopolysaccharide (LPS) or by the administration of l-arginine in wild-type or XIAP-deficient mice, and ex vivo model was induced by the administration of cerulein+LPS in AR42J cell line following XIAP inhibition. The severity of acute pancreatitis was determined by serum amylase activity and histological grading. XIAP deletion on cell apoptosis, necrosis and inflammatory response were examined. Caspases activities, nuclear factor-κB (NF-κB) activation and receptor-interacting protein kinase1 (RIP1) degradation were assessed by western blot. Deletion of XIAP resulted in the reduction of amylase activity, decrease of NF-κB activation and less release of TNF-α and IL-6, together with increased caspases activities and RIP1 degradation, leading to enhanced apoptosis and reduced necrosis in pancreatic acinar cells and ameliorated the severity of acute pancreatitis. Our results indicate that deletion of XIAP switches cell death away from necrosis to apoptosis and decreases the inflammatory response, effectively attenuating the severity of AP/SAP. The critical role of XIAP in cell death and inflammation suggests that inhibition of XIAP represents a potential therapeutic strategy for the treatment of acute pancreatitis.
Collapse
|
94
|
Derakhshan A, Chen Z, Van Waes C. Therapeutic Small Molecules Target Inhibitor of Apoptosis Proteins in Cancers with Deregulation of Extrinsic and Intrinsic Cell Death Pathways. Clin Cancer Res 2017; 23:1379-1387. [PMID: 28039268 PMCID: PMC5354945 DOI: 10.1158/1078-0432.ccr-16-2172] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023]
Abstract
The Cancer Genome Atlas (TCGA) has unveiled genomic deregulation of various components of the extrinsic and intrinsic apoptotic pathways in different types of cancers. Such alterations are particularly common in head and neck squamous cell carcinomas (HNSCC), which frequently display amplification and overexpression of the Fas-associated via death domain (FADD) and inhibitor of apoptosis proteins (IAP) that complex with members of the TNF receptor family. Second mitochondria-derived activator of caspases (SMAC) mimetics, modeled after the endogenous IAP antagonist SMAC, and IAP inhibitors represent important classes of novel small molecules currently in phase I/II clinical trials. Here we review the physiologic roles of IAPs, FADD, and other components involved in cell death, cell survival, and NF-κB signaling pathways in cancers, including HNSCC. We summarize the results of targeting IAPs in preclinical models of HNSCC using SMAC mimetics. Synergistic activity of SMAC mimetics together with death agonists TNFα or TRAIL occurred in vitro, whereas their antitumor effects were augmented when combined with radiation and chemotherapeutic agents that induce TNFα in vivo In addition, clinical trials testing SMAC mimetics as single agents or together with chemo- or radiation therapies in patients with HNSCC and solid tumors are summarized. As we achieve a deeper understanding of the genomic alterations and molecular mechanisms underlying deregulated death and survival pathways in different cancers, the role of SMAC mimetics and IAP inhibitors in cancer treatment will be elucidated. Such developments could enhance precision therapeutics and improve outcomes for cancer patients. Clin Cancer Res; 23(6); 1379-87. ©2016 AACR.
Collapse
Affiliation(s)
- Adeeb Derakhshan
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
95
|
Modulation of ARTS and XIAP by Parkin Is Associated with Carnosic Acid Protects SH-SY5Y Cells against 6-Hydroxydopamine-Induced Apoptosis. Mol Neurobiol 2017; 55:1786-1794. [DOI: 10.1007/s12035-017-0443-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/03/2017] [Indexed: 01/27/2023]
|
96
|
|
97
|
Shakeri R, Kheirollahi A, Davoodi J. Apaf-1: Regulation and function in cell death. Biochimie 2017; 135:111-125. [PMID: 28192157 DOI: 10.1016/j.biochi.2017.02.001] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/08/2023]
Abstract
Apoptosis, a form of programmed cell death, is responsible for eliminating damaged or unnecessary cells in multicellular organisms. Various types of intracellular stress trigger apoptosis by induction of cytochrome c release from mitochondria into the cytosol. Apoptotic protease activating factor-1 (Apaf-1) is a key molecule in the intrinsic or mitochondrial pathway of apoptosis, which oligomerizes in response to cytochrome c release and forms a large complex known as apoptosome. Procaspase-9, an initiator caspase in the mitochondrial pathway, is recruited and activated by the apoptosome leading to downstream caspase-3 processing. Various cellular proteins and small molecules can modulate apoptosome formation and function directly or indirectly. Despite recent progress in understanding the mitochondrial pathway of apoptosis, numerous questions such as the molecular mechanism of Apaf-1 oligomerization and caspase-9 activation remain poorly understood. In addition, reports have emerged showing non-apoptotic functions for Apaf-1. The current review summarizes the latest findings regarding structure-function relationship of Apaf-1 as well as its modifiers.
Collapse
Affiliation(s)
- Raheleh Shakeri
- Department of Biological Science and Biotechnology, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Asma Kheirollahi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Jamshid Davoodi
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran.
| |
Collapse
|
98
|
Tang HM, Talbot CC, Fung MC, Tang HL. Molecular signature of anastasis for reversal of apoptosis. F1000Res 2017; 6:43. [PMID: 28299189 DOI: 10.12688/f1000research.10568.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis in vitro and in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis - the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
99
|
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis
in vitro and
in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis – the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
100
|
Zumbrägel FK, Machtens DA, Curth U, Lüder CG, Reubold TF, Eschenburg S. Survivin does not influence the anti-apoptotic action of XIAP on caspase-9. Biochem Biophys Res Commun 2017; 482:530-535. [DOI: 10.1016/j.bbrc.2016.11.094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 10/20/2022]
|