1
|
Wang J, Zhao R, Liu Y, Hu T, Li X, He L, Guo Z, Chen C, Shi X. The correlation between Smac, IAPs and mitochondrial apoptosis, muscle tenderness during postmortem aging of Oula Tibetan sheep meat. Food Chem X 2024; 24:101887. [PMID: 39498258 PMCID: PMC11532436 DOI: 10.1016/j.fochx.2024.101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
Oula Tibetan sheep meat has rich nutritional value but relatively poor tenderness. Recently, apoptosis of muscle cells has gradually become a research hotspot for improving meat tenderness during postmortem aging. Smac can promote the decrease of IAPs expression in tumor cells, thereby inducing mitochondrial apoptosis. However, the relationship between Smac, IAPs and mitochondrial apoptosis, muscle tenderness during postmortem meat aging is still unclear. Thus, the aim of this work was to explore the relationship between Smac, IAPs and mitochondrial apoptosis as well as muscle tenderness during postmortem meat aging. Smac concentration, IAPs concentration, pH value, ATP content, SDH activity, MPTP opening degree, MMP, caspase-3/9 activity, apoptotic rate, MFI and shear force value of Oula Tibetan sheep meat were measured at different aging times and correlation analysis was performed. Correlation analysis revealed that Smac, IAPs were markedly related to mitochondrial apoptosis and muscle tenderness during postmortem aging of Tibetan sheep meat. The results suggest that Smac may regulate IAPs to promote mitochondrial apoptosis and muscle tenderization in Oula Tibetan sheep meat during postmortem aging.
Collapse
Affiliation(s)
- Jingyu Wang
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Ruina Zhao
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Yang Liu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Tieying Hu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaolong Li
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Long He
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhaobin Guo
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Cheng Chen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - Xixiong Shi
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
2
|
Schorn F, Werthenbach JP, Hoffmann M, Daoud M, Stachelscheid J, Schiffmann LM, Hildebrandt X, Lyu SI, Peltzer N, Quaas A, Vucic D, Silke J, Pasparakis M, Kashkar H. cIAPs control RIPK1 kinase activity-dependent and -independent cell death and tissue inflammation. EMBO J 2023; 42:e113614. [PMID: 37789765 PMCID: PMC10646551 DOI: 10.15252/embj.2023113614] [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: 01/27/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Cellular inhibitor of apoptosis proteins (cIAPs) are RING-containing E3 ubiquitin ligases that ubiquitylate receptor-interacting protein kinase 1 (RIPK1) to regulate TNF signalling. Here, we established mice simultaneously expressing enzymatically inactive cIAP1/2 variants, bearing mutations in the RING domains of cIAP1/2 (cIAP1/2 mutant RING, cIAP1/2MutR ). cIap1/2MutR/MutR mice died during embryonic development due to RIPK1-mediated apoptosis. While expression of kinase-inactive RIPK1D138N rescued embryonic development, Ripk1D138N/D138N /cIap1/2MutR/MutR mice developed systemic inflammation and died postweaning. Cells expressing cIAP1/2MutR and RIPK1D138N were still susceptible to TNF-induced apoptosis and necroptosis, implying additional kinase-independent RIPK1 activities in regulating TNF signalling. Although further ablation of Ripk3 did not lead to any phenotypic improvement, Tnfr1 gene knock-out prevented early onset of systemic inflammation and premature mortality, indicating that cIAPs control TNFR1-mediated toxicity independent of RIPK1 and RIPK3. Beyond providing novel molecular insights into TNF-signalling, the mouse model established in this study can serve as a useful tool to further evaluate ongoing therapeutic protocols using inhibitors of TNF, cIAPs and RIPK1.
Collapse
Affiliation(s)
- Fabian Schorn
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - J Paul Werthenbach
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Mattes Hoffmann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Mila Daoud
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Johanna Stachelscheid
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
| | - Lars M Schiffmann
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral, Cancer and Transplantation SurgeryUniversity of CologneCologneGermany
| | - Ximena Hildebrandt
- Faculty of Medicine and University Hospital of Cologne, Department of Translational GenomicsUniversity of CologneCologneGermany
| | - Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology and Center for Integrated Oncology (CIO) Cologne BonnUniversity of CologneCologneGermany
| | - Nieves Peltzer
- Faculty of Medicine and University Hospital of Cologne, Department of Translational GenomicsUniversity of CologneCologneGermany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology and Center for Integrated Oncology (CIO) Cologne BonnUniversity of CologneCologneGermany
| | - Domagoj Vucic
- Department of Immunology DiscoveryGenentechSouth San FranciscoCAUSA
| | - John Silke
- The Walter and Eliza Hall Institute for Medical ResearchMelbourneVic.Australia
| | - Manolis Pasparakis
- Institute for GeneticsUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Hamid Kashkar
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular ImmunologyUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| |
Collapse
|
3
|
Ge L, Wang T, Shi D, Geng Y, Fan H, Zhang R, Zhang Y, Zhao J, Li S, Li Y, Shi H, Song G, Pan J, Wang L, Han J. ATF6α contributes to rheumatoid arthritis by inducing inflammatory cytokine production and apoptosis resistance. Front Immunol 2022; 13:965708. [PMID: 36300114 PMCID: PMC9590309 DOI: 10.3389/fimmu.2022.965708] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Objective The contribution of activating transcription factor 6α (ATF6α) in rheumatoid arthritis (RA) pathogenesis, especially on fibroblast-like synoviocytes (FLSs), has been suggested by its sensitivity to inflammatory stimulus. However, the exact role and therapeutic potential of ATF6α in RA remains to be fully elucidated. Methods ATF6α expression was determined in joint tissues and FLS, and gain-of-function and loss-of-function analyses were applied to evaluate the biological roles of ATF6α in RA FLSs. A murine collagen-induced arthritis (CIA) model, combining both gene deletion of ATF6α and treatment with the ATF6α inhibitor Ceapin-A7, was employed. Joint inflammation, tissue destruction, circulating levels of inflammatory cytokines were assessed in CIA mice. Transcriptome sequencing analysis (RNASeq), molecular biology, and biochemical approaches were performed to identify target genes of ATF6α. Results ATF6α expression was significantly increased in synovium of RA patients and in synovium of mice subjected to CIA. ATF6α silencing or inhibition repressed RA FLSs viability and cytokine production but induced the apoptosis. CIA-model mice with ATF6α deficiency displayed decreased arthritic progression, leading to profound reductions in clinical and proinflammatory markers in the joints. Pharmacological treatment of mice with Ceapin-A7 reduced arthritis severity in CIA models. RNA-sequencing of wild-type and knockdown of ATF6α in RA FLSs revealed a transcriptional program that promotes inflammation and suppresses apoptosis, and subsequent experiments identified Baculoviral IAP Repeat Containing 3 (BIRC3) as the direct target for ATF6α. Conclusion This study highlights the pathogenic role of ATF6α-BIRC3 axis in RA and identifies a novel pathway for new therapies against RA.
Collapse
Affiliation(s)
- Luna Ge
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Ting Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Dandan Shi
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Yun Geng
- Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, China
| | - Huancai Fan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Yuang Zhang
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Jianli Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
| | - Yi Li
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University (Shandong Provincial Hospital), Jinan, China
| | - Haojun Shi
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, China
| | - Jihong Pan
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
| | - Lin Wang
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
- *Correspondence: Lin Wang, ; Jinxiang Han,
| | - Jinxiang Han
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji’nan, China
- *Correspondence: Lin Wang, ; Jinxiang Han,
| |
Collapse
|
4
|
Witkop EM, Proestou DA, Gomez-Chiarri M. The expanded inhibitor of apoptosis gene family in oysters possesses novel domain architectures and may play diverse roles in apoptosis following immune challenge. BMC Genomics 2022; 23:201. [PMID: 35279090 PMCID: PMC8917759 DOI: 10.1186/s12864-021-08233-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background Apoptosis plays important roles in a variety of functions, including immunity and response to environmental stress. The Inhibitor of Apoptosis (IAP) gene family of apoptosis regulators is expanded in molluscs, including eastern, Crassostrea virginica, and Pacific, Crassostrea gigas, oysters. The functional importance of IAP expansion in apoptosis and immunity in oysters remains unknown. Results Phylogenetic analysis of IAP genes in 10 molluscs identified lineage specific gene expansion in bivalve species. Greater IAP gene family expansion was observed in C. virginica than C. gigas (69 vs. 40), resulting mainly from tandem duplications. Functional domain analysis of oyster IAP proteins revealed 3 novel Baculoviral IAP Repeat (BIR) domain types and 14 domain architecture types across gene clusters, 4 of which are not present in model organisms. Phylogenetic analysis of bivalve IAPs suggests a complex history of domain loss and gain. Most IAP genes in oysters (76% of C. virginica and 82% of C. gigas), representing all domain architecture types, were expressed in response to immune challenge (Ostreid Herpesvirus OsHV-1, bacterial probionts Phaeobacter inhibens and Bacillus pumilus, several Vibrio spp., pathogenic Aliiroseovarius crassostreae, and protozoan parasite Perkinsus marinus). Patterns of IAP and apoptosis-related differential gene expression differed between the two oyster species, where C. virginica, in general, differentially expressed a unique set of IAP genes in each challenge, while C. gigas differentially expressed an overlapping set of IAP genes across challenges. Apoptosis gene expression patterns clustered mainly by resistance/susceptibility of the oyster host to immune challenge. Weighted Gene Correlation Network Analysis (WGCNA) revealed unique combinations of transcripts for 1 to 12 IAP domain architecture types, including novel types, were significantly co-expressed in response to immune challenge with transcripts in apoptosis-related pathways. Conclusions Unprecedented diversity characterized by novel BIR domains and protein domain architectures was observed in oyster IAPs. Complex patterns of gene expression of novel and conserved IAPs in response to a variety of ecologically-relevant immune challenges, combined with evidence of direct co-expression of IAP genes with apoptosis-related transcripts, suggests IAP expansion facilitates complex and nuanced regulation of apoptosis and other immune responses in oysters. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08233-6.
Collapse
|
5
|
Sørensen MD, Kristensen BW. TUMOUR-ASSOCIATED CD204+ MICROGLIA/MACROPHAGES ACCUMULATE IN PERIVASCULAR AND PERINECROTIC NICHES AND CORRELATE WITH AN INTERLEUKIN-6 ENRICHED INFLAMMATORY PROFILE IN GLIOBLASTOMA. Neuropathol Appl Neurobiol 2021; 48:e12772. [PMID: 34713474 PMCID: PMC9306597 DOI: 10.1111/nan.12772] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Mia Dahl Sørensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Li S, Zheng X, Hu Y, You K, Wang J. RNF31 mediated ubiquitination of A20 aggravates inflammation and hepatocyte apoptosis through the TLR4/MyD88/NF-κB signaling pathway. Chem Biol Interact 2021; 348:109623. [PMID: 34416243 DOI: 10.1016/j.cbi.2021.109623] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/16/2021] [Indexed: 01/06/2023]
Abstract
Inflammatory cytokine storm is one of the main pathogenesis of acute liver injury, and accumulating evidence suggests that the E3 ubiquitin ligase ring finger protein 31 (RNF31) plays an important regulatory role in the activation of inflammatory pathways. We found that RNF31 expression was up-regulated in lipopolysaccharide (LPS)-treated HL-7702 cells. Western blotting results showed decreased expression of RNF31 and total ubiquitinated proteins after transfection of si-RNF31. The results of MTT assay indicated that cell viability was enhanced. Flow cytometry analysis showed that cell apoptosis and ROS content was decreased, and ELISA assay results exhibited that the inflammatory factors secretion was reduced. Interestingly, A20 protein expression was inhibited as RNF31 expression was upregulated. On this basis, we performed co-immunoprecipitation assays and found that RNF31 could interact with A20. Actinomycin tracing and proteasome inhibition experiments showed that RNF31 degrades A20 through the proteasome pathway. Furthermore, overexpression of A20 enhanced cell viability, reduced apoptosis, and inhibited ROS generation and inflammatory factor secretion. Mechanistic studies revealed that RNF31 was able to degrade A20, which affected the inflammatory response and hepatocyte apoptosis mediated by the toll like receptor 4 (TLR4)/myeloid differentiation factor88 (MyD88)/nuclear transcription factor-κB (NF-κB) signaling pathway. Moreover, knockdown of RNF31 attenuated the inflammatory response induced by d-Gal/LPS in mice with acute liver injury. In conclusion, RNF31 degrades A20 by ubiquitination and activates the TLR4/MyD88/NF-κB signaling pathway to aggravate acute liver injury.
Collapse
Affiliation(s)
- Song Li
- Department of laboratory medicine, Zhumadian Central Hospital, Zhumadian, 463000, Henan, China.
| | - Ximing Zheng
- Department of laboratory medicine, Zhumadian Central Hospital, Zhumadian, 463000, Henan, China
| | - Yingchao Hu
- Department of laboratory medicine, Zhumadian Central Hospital, Zhumadian, 463000, Henan, China
| | - Kun You
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Junda Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| |
Collapse
|
7
|
den Besten W, Verma K, Yamazoe S, Blaquiere N, Phung W, Izrael-Tomasevic A, Mulvihill MM, Helgason E, Prakash S, Goncharov T, Vucic D, Dueber E, Fairbrother WJ, Wertz I, Yu K, Staben ST. Primary Amine Tethered Small Molecules Promote the Degradation of X-Linked Inhibitor of Apoptosis Protein. J Am Chem Soc 2021; 143:10571-10575. [PMID: 34236858 DOI: 10.1021/jacs.1c05269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We hypothesized that the proximity-driven ubiquitylation of E3-interacting small molecules could affect the degradation of E3 ubiquitin ligases. A series of XIAP BIR2 domain-binding small molecules was modified to append a nucleophilic primary amine. This modification transforms XIAP binders into inducers of XIAP degradation. The degradation of XIAP is E1- and proteasome-dependent, dependent on the ligase function of XIAP, and is rescued by subtle modifications of the small molecule that would obviate ubiquitylation. We demonstrate in vitro ubiquitylation of the small molecule that is dependent on its interaction with XIAP. Taken together, these results demonstrate the designed ubiquitylation of an engineered small molecule and a novel approach for the degradation of E3 ubiquitin ligases.
Collapse
|
8
|
He L, Sehrawat TS, Verma VK, Navarro-Corcuera A, Sidhu G, Mauer A, Luo X, Katsumi T, Chen J, Shah S, Arab JP, Cao S, Kashkar H, Gores GJ, Malhi H, Shah VH. XIAP Knockdown in Alcohol-Associated Liver Disease Models Exhibits Divergent in vitro and in vivo Phenotypes Owing to a Potential Zonal Inhibitory Role of SMAC. Front Physiol 2021; 12:664222. [PMID: 34025452 PMCID: PMC8138467 DOI: 10.3389/fphys.2021.664222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol-associated liver disease (ALD) has been recognized as the most common cause of advanced liver disease worldwide, though mechanisms of pathogenesis remain incompletely understood. The X-linked inhibitor of apoptosis (XIAP) protein was originally described as an anti-apoptotic protein that directly binds and inhibits caspases-3, 7, and 9. Here, we investigated the function of XIAP in hepatocytes in vitro using gain and loss-of-function approaches. We noted an XIAP-dependent increase in caspase activation as well as increased inflammatory markers and pro-inflammatory EV release from hepatocytes in vitro. Primary hepatocytes (PMH) from Xiap Alb.Cre and Xiap loxP mice exhibited higher cell death but surprisingly, lower expression of inflammation markers. Conditioned media from these isolated Xiap deleted PMH further decrease inflammation in bone marrow-derived macrophages. Also, interestingly, when administered an ethanol plus Fas-agonist-Jo2 model and an ethanol plus CCl4 model, these animals failed to develop an exacerbated disease phenotype in vivo. Of note, neither Xiap Alb . Cre nor Xiap AAV8.Cre mice presented with aggravated liver injury, hepatocyte apoptosis, liver steatosis, or fibrosis. Since therapeutics targeting XIAP are currently in clinical trials and caspase-induced death is very important for development of ALD, we sought to explore the potential basis of this unexpected lack of effect. We utilized scRNA-seq and spatially reconstructed hepatocyte transcriptome data from human liver tissue and observed that XIAP was significantly zonated, along with its endogenous inhibitor second mitochondria-derived activator of caspases (SMAC) in periportal region. This contrasted with pericentral zonation of other IAPs including cIAP1 and Apollon as well as caspases 3, 7, and 9. Thus providing a potential explanation for compensation of the effect of Xiap deletion by other IAPs. In conclusion, our findings implicate a potential zonallydependent role for SMAC that prevented development of a phenotype in XIAP knockout mice in ALD models. Targeting SMAC may also be important in addition to current efforts of targeting XIAP in treatment of ALD.
Collapse
Affiliation(s)
- Li He
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tejasav S. Sehrawat
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Vikas K. Verma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Amaia Navarro-Corcuera
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Guneet Sidhu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Amy Mauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xin Luo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tomohiro Katsumi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jingbiao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Soni Shah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Juan Pablo Arab
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Sheng Cao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hamid Kashkar
- Centre for Molecular Medicine Cologne and Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
9
|
Developmental Attenuation of Neuronal Apoptosis by Neural-Specific Splicing of Bak1 Microexon. Neuron 2020; 107:1180-1196.e8. [PMID: 32710818 DOI: 10.1016/j.neuron.2020.06.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/29/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022]
Abstract
Continuous neuronal survival is vital for mammals because mammalian brains have limited regeneration capability. After neurogenesis, suppression of apoptosis is needed to ensure a neuron's long-term survival. Here we describe a robust genetic program that intrinsically attenuates apoptosis competence in neurons. Developmental downregulation of the splicing regulator PTBP1 in immature neurons allows neural-specific splicing of the evolutionarily conserved Bak1 microexon 5. Exon 5 inclusion triggers nonsense-mediated mRNA decay (NMD) and unproductive translation of Bak1 transcripts (N-Bak mRNA), leading to suppression of pro-apoptotic BAK1 proteins and allowing neurons to reduce apoptosis. Germline heterozygous ablation of exon 5 increases BAK1 proteins exclusively in the brain, inflates neuronal apoptosis, and leads to early postnatal mortality. Therefore, neural-specific exon 5 splicing and depletion of BAK1 proteins uniquely repress neuronal apoptosis. Although apoptosis is important for development, attenuation of apoptosis competence through neural-specific splicing of the Bak1 microexon is essential for neuronal and animal survival.
Collapse
|
10
|
Zhang J, Webster JD, Dugger DL, Goncharov T, Roose-Girma M, Hung J, Kwon YC, Vucic D, Newton K, Dixit VM. Ubiquitin Ligases cIAP1 and cIAP2 Limit Cell Death to Prevent Inflammation. Cell Rep 2020; 27:2679-2689.e3. [PMID: 31141691 DOI: 10.1016/j.celrep.2019.04.111] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/29/2019] [Accepted: 04/26/2019] [Indexed: 01/18/2023] Open
Abstract
Cellular inhibitor of apoptosis proteins cIAP1 and cIAP2 ubiquitinate nuclear factor κB (NF-κB)-inducing kinase (NIK) to suppress non-canonical NF-κB signaling and substrates such as receptor interacting protein kinase 1 (RIPK1) to promote cell survival. We investigate how these functions contribute to homeostasis by eliminating cIap2 from adult cIap1-deficient mice. cIAP1 and cIAP2 (cIAP1/2) deficiency causes rapid weight loss and inflammation, with aberrant cell death, indicated by cleaved caspases-3 and -8, prevalent in intestine and liver. Deletion of Casp8 and Ripk3 prevents this aberrant cell death, reduces the inflammation, and prolongs mouse survival, whereas Ripk3 loss alone offers little benefit. Residual inflammation in mice lacking cIap1/2, Casp8, and Ripk3 is reduced by inhibition of NIK. Loss of Casp8 and Mlkl (mixed lineage kinase domain-like), but not Mlkl loss alone, also prevents cIAP1/2-deficient mice from dying around embryonic day 11. Therefore, a major function of cIAP1/2 in vivo is to suppress caspase-8-dependent cell death.
Collapse
Affiliation(s)
- Jieqiong Zhang
- Department of Physiological Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Joshua D Webster
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Debra L Dugger
- Department of Physiological Chemistry, Genentech, South San Francisco, CA 94080, USA
| | - Tatiana Goncharov
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, South San Francisco, CA 94080, USA
| | - Jeffrey Hung
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Youngsu C Kwon
- Department of Translational Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA 94080, USA
| | - Kim Newton
- Department of Physiological Chemistry, Genentech, South San Francisco, CA 94080, USA.
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
11
|
Wang W, Ji Y, Yang W, Zhang C, Angwa L, Jin B, Liu J, Lv M, Ma W, Yang J, Wang K. Inhibitors of apoptosis proteins (IAPs) are associated with T-2 toxin-induced decreased collagen II in mouse chondrocytes in vitro. Toxicon 2020; 176:34-43. [PMID: 32103793 DOI: 10.1016/j.toxicon.2020.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/14/2019] [Accepted: 01/09/2020] [Indexed: 10/25/2022]
Abstract
T-2 toxin is considered an unavoidable pollutant, which contaminates food crops and stockpiled cereals, impairing the health of humans and animals due to its multi-organ toxicity. Studies have shown that T-2 toxin can cause articular cartilage damage; however, the underlying molecular mechanism is still unclear. Here, we investigated the possible mechanism of the following inhibitors of apoptosis proteins (IAPs) family members: NAIP, cIAP1, cIAP2, XIAP, and Survivin, and their involvement in T-2 toxin-induced mouse chondrocyte damage. In this study, mouse articular chondrocytes were isolated and cultured in vitro, and the chondrocytes were then treated with 0, 5, 10, and 20 ng/mL T-2 toxin. Firstly, the toxic effect of T-2 toxin on chondrocytes was determined. CCK-8 assay results showed that T-2 toxin induced a dose-dependent inhibition of chondrocyte viability. Transmission electron microscopy demonstrated that T-2 toxin caused morphological changes in chondrocyte endoplasmic reticulum and an increase in mitochondrial swelling. In addition, Annexin-V-FITC/PI staining and caspase 3 protein expression showed that T-2 toxin induced an increase in the apoptotic rate of chondrocytes. Secondly, it was found that T-2 toxin cause decreased expression of cellular and secreted Collagen II. Finally, we examined the expression of NAIP, cIAP1, cIAP2, XIAP, and Survivin in chondrocytes in the presence of T-2 toxin and their relationship with decreased Collagen II. The decrease in Collagen II was negatively correlated with the expression of cIAP1, cIAP2 and positively correlated with NAIP and Survivin mRNA level. Survivin mRNA level had a positive correlation with Collagen II as shown by partial correlation analysis. This study revealed the new role of IAPs in chondrocyte injury and provides new insights and clues into the mechanism of T-2 toxin-induced chondrocyte damage.
Collapse
Affiliation(s)
- Wenji Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Yi Ji
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Wenjing Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Chengzhi Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China
| | - Linet Angwa
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Department of Clinical Medicine, Kabarak University, Private Bag, 20157, Kabarak, Kenya
| | - Baiming Jin
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China; School of Public Health, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - Juan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Man Lv
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Wenjing Ma
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Jie Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China
| | - Kewei Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention/ Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Institute of Cell Biotechnology, China and Russia Medical Research Center, Harbin Medical University, Harbin, 150081, China; Departments of Surgery, University of Illinois College of Medicine, One Illini Drive, Peoria, IL, 61605, USA.
| |
Collapse
|
12
|
Lala-Tabbert N, Lejmi-Mrad R, Timusk K, Fukano M, Holbrook J, St-Jean M, LaCasse EC, Korneluk RG. Targeted ablation of the cellular inhibitor of apoptosis 1 (cIAP1) attenuates denervation-induced skeletal muscle atrophy. Skelet Muscle 2019; 9:13. [PMID: 31126323 PMCID: PMC6533726 DOI: 10.1186/s13395-019-0201-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/13/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Skeletal muscle atrophy is a pathological condition that contributes to morbidity in a variety of conditions including denervation, cachexia, and aging. Muscle atrophy is characterized as decreased muscle fiber cross-sectional area and protein content due, in part, to the proteolytic activities of two muscle-specific E3 ubiquitin ligases: muscle RING-finger 1 (MuRF1) and muscle atrophy F-box (MAFbx or Atrogin-1). The nuclear factor-kappa B (NF-κB) pathway has emerged as a critical signaling network in skeletal muscle atrophy and has become a prime therapeutic target for the treatment of muscle diseases. Unfortunately, none of the NF-κB targeting drugs are currently being used to treat these diseases, likely because of our limited knowledge and specificity, for muscle biology and disease. The cellular inhibitor of apoptosis 1 (cIAP1) protein is a positive regulator of tumor necrosis factor alpha (TNFα)-mediated classical NF-κB signaling, and cIAP1 loss has been shown to enhance muscle regeneration during acute and chronic injury. METHODS Sciatic nerve transection in wild-type, cIAP1-null and Smac mimetic compound (SMC)-treated mice was performed to investigate the role of cIAP1 in denervation-induced atrophy. Genetic in vitro models of C2C12 myoblasts and primary myoblasts were also used to examine the role of classical NF-κB activity in cIAP1-induced myotube atrophy. RESULTS We found that cIAP1 expression was upregulated in denervated muscles compared to non-denervated controls 14 days after denervation. Genetic and pharmacological loss of cIAP1 attenuated denervation-induced muscle atrophy and overexpression of cIAP1 in myotubes was sufficient to induce atrophy. The induction of myotube atrophy by cIAP1 was attenuated when the classical NF-κB signaling pathway was inhibited. CONCLUSIONS These results demonstrate the cIAP1 is an important mediator of NF-κB/MuRF1 signaling in skeletal muscle atrophy and is a promising therapeutic target for muscle wasting diseases.
Collapse
Affiliation(s)
- Neena Lala-Tabbert
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Rim Lejmi-Mrad
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Kristen Timusk
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Marina Fukano
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Janelle Holbrook
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Martine St-Jean
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Eric C LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Robert G Korneluk
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
13
|
Jeong Y, Lim JW, Kim H. Lycopene Inhibits Reactive Oxygen Species-Mediated NF-κB Signaling and Induces Apoptosis in Pancreatic Cancer Cells. Nutrients 2019; 11:nu11040762. [PMID: 30939781 PMCID: PMC6521322 DOI: 10.3390/nu11040762] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 12/21/2022] Open
Abstract
Generation of excess quantities of reactive oxygen species (ROS) caused by mitochondrial dysfunction facilitates rapid growth of pancreatic cancer cells. Elevated ROS levels in cancer cells cause an anti-apoptotic effect by activating survival signaling pathways, such as NF-κB and its target gene expression. Lycopene, a carotenoid found in tomatoes and a potent antioxidant, displays a protective effect against pancreatic cancer. The present study was designed to determine if lycopene induces apoptosis of pancreatic cancer PANC-1 cells by decreasing intracellular and mitochondrial ROS levels, and consequently suppressing NF-κB activation and expression of NF-κB target genes including cIAP1, cIAP2, and survivin. The results show that the lycopene decreased intracellular and mitochondrial ROS levels, mitochondrial function (determined by the mitochondrial membrane potential and oxygen consumption rate), NF-κB activity, and expression of NF-κB-dependent survival genes in PANC-1 cells. Lycopene reduced cell viability with increases in active caspase-3 and the Bax to Bcl-2 ratio in PANC-1 cells. These findings suggest that supplementation of lycopene could potentially reduce the incidence of pancreatic cancer.
Collapse
Affiliation(s)
- Yoonseon Jeong
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Joo Weon Lim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|
14
|
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
|
15
|
Annibaldi A, Wicky John S, Vanden Berghe T, Swatek KN, Ruan J, Liccardi G, Bianchi K, Elliott PR, Choi SM, Van Coillie S, Bertin J, Wu H, Komander D, Vandenabeele P, Silke J, Meier P. Ubiquitin-Mediated Regulation of RIPK1 Kinase Activity Independent of IKK and MK2. Mol Cell 2019; 69:566-580.e5. [PMID: 29452637 PMCID: PMC5823975 DOI: 10.1016/j.molcel.2018.01.027] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 12/11/2017] [Accepted: 01/19/2018] [Indexed: 10/25/2022]
Abstract
Tumor necrosis factor (TNF) can drive inflammation, cell survival, and death. While ubiquitylation-, phosphorylation-, and nuclear factor κB (NF-κB)-dependent checkpoints suppress the cytotoxic potential of TNF, it remains unclear whether ubiquitylation can directly repress TNF-induced death. Here, we show that ubiquitylation regulates RIPK1's cytotoxic potential not only via activation of downstream kinases and NF-kB transcriptional responses, but also by directly repressing RIPK1 kinase activity via ubiquitin-dependent inactivation. We find that the ubiquitin-associated (UBA) domain of cellular inhibitor of apoptosis (cIAP)1 is required for optimal ubiquitin-lysine occupancy and K48 ubiquitylation of RIPK1. Independently of IKK and MK2, cIAP1-mediated and UBA-assisted ubiquitylation suppresses RIPK1 kinase auto-activation and, in addition, marks it for proteasomal degradation. In the absence of a functional UBA domain of cIAP1, more active RIPK1 kinase accumulates in response to TNF, causing RIPK1 kinase-mediated cell death and systemic inflammatory response syndrome. These results reveal a direct role for cIAP-mediated ubiquitylation in controlling RIPK1 kinase activity and preventing TNF-mediated cytotoxicity.
Collapse
Affiliation(s)
- Alessandro Annibaldi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
| | - Sidonie Wicky John
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Tom Vanden Berghe
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kirby N Swatek
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Jianbin Ruan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Room 3024B, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Gianmaria Liccardi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Katiuscia Bianchi
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK; Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Paul R Elliott
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Sze Men Choi
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Samya Van Coillie
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - John Bertin
- Pattern Recognition Receptor DPU and Platform Technology and Science, GlaxoSmithKline, Collegeville Road, Collegeville, PA 19426, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Room 3024B, 3 Blackfan Circle, Boston, MA 02115, USA
| | - David Komander
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Peter Vandenabeele
- VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
| |
Collapse
|
16
|
Abstract
The inhibitor of apoptosis proteins (IAPs) are a family of proteins that were chiefly known for their ability to inhibit apoptosis by blocking caspase activation or activity. Recent research has shown that cellular IAP1 (cIAP1), cIAP2, and X-linked IAP (XIAP) also regulate signaling by receptors of the innate immune system by ubiquitylating their substrates. These IAPs thereby act at the intersection of pathways leading to cell death and inflammation. Mutation of IAP genes can impair tissue homeostasis and is linked to several human diseases. Small-molecule IAP antagonists have been developed to treat certain malignant, infectious, and inflammatory diseases. Here, we will discuss recent advances in our understanding of the functions of cIAP1, cIAP2, and XIAP; the consequences of their mutation or dysregulation; and the therapeutic potential of IAP antagonist drugs.
Collapse
Affiliation(s)
- Najoua Lalaoui
- Cell Signalling and Cell Death, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3050, Australia
| | - David Lawrence Vaux
- Cell Signalling and Cell Death, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3050, Australia
| |
Collapse
|
17
|
Seo J, Kim MW, Bae KH, Lee SC, Song J, Lee EW. The roles of ubiquitination in extrinsic cell death pathways and its implications for therapeutics. Biochem Pharmacol 2018; 162:21-40. [PMID: 30452908 DOI: 10.1016/j.bcp.2018.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/14/2018] [Indexed: 01/24/2023]
Abstract
Regulation of cell survival and death, including apoptosis and necroptosis, is important for normal development and tissue homeostasis, and disruption of these processes can cause cancer, inflammatory diseases, and degenerative diseases. Ubiquitination is a cellular process that induces proteasomal degradation by covalently attaching ubiquitin to the substrate protein. In addition to proteolytic ubiquitination, nonproteolytic ubiquitination, such as M1-linked and K63-linked ubiquitination, has been shown to be important in recent studies, which have demonstrated its function in cell signaling pathways that regulate inflammation and cell death pathways. In this review, we summarize the TRAIL- and TNF-induced death receptor signaling pathways along with recent advances in this field and illustrate how different types of ubiquitination control cell death and survival. In particular, we provide an overview of the different types of ubiquitination, target residues, and modifying enzymes, including E3 ligases and deubiquitinating enzymes. Given the relevance of these regulatory pathways in human disease, we hope that a better understanding of the regulatory mechanisms of cell death pathways will provide insights into and therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Jinho Seo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Min Wook Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Republic of Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.
| |
Collapse
|
18
|
|
19
|
Abstract
Inhibitor of apoptosis (IAP) family comprises a group of endogenous proteins that function as main regulators of caspase activity and cell death. They are considered the main culprits in evasion of apoptosis, which is a fundamental hallmark of carcinogenesis. Overexpression of IAP proteins has been documented in various solid and hematological malignancies, rendering them resistant to standard chemotherapeutics and radiation therapy and conferring poor prognosis. This observation has urged their exploitation as therapeutic targets in cancer with promising pre-clinical outcomes. This review describes the structural and functional features of IAP proteins to elucidate the mechanism of their anti-apoptotic activity. We also provide an update on patterns of IAP expression in different tumors, their impact on treatment response and prognosis, as well as the emerging investigational drugs targeting them. This aims at shedding the light on the advances in IAP targeting achieved to date, and encourage further development of clinically applicable therapeutic approaches.
Collapse
Affiliation(s)
- Mervat S Mohamed
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Kingdom of Saudi Arabia.
- Department of Chemistry, Biochemistry Speciality, Faculty of Science, Cairo University, Giza, Egypt.
- , Tabuk, Kingdom of Saudi Arabia.
| | - Mai K Bishr
- Department of Radiotherapy, Children's Cancer Hospital Egypt (CCHE), Cairo, Egypt
| | - Fahad M Almutairi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Ayat G Ali
- Department of Biochemistry, El Sahel Teaching Hospital, Cairo, Egypt
| |
Collapse
|
20
|
Dougan SK, Dougan M. Regulation of innate and adaptive antitumor immunity by IAP antagonists. Immunotherapy 2018; 10:787-796. [PMID: 29807457 DOI: 10.2217/imt-2017-0185] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Inhibition of the T-cell co-inhibitory checkpoint receptors or their ligands CTLA-4, PD-1 and PD-L1 using monoclonal antibodies has proven to be highly effective against many cancers. Yet many cancers remain resistant to checkpoint blockade, and durable remissions occur in only a minority of patients. Novel approaches to enhancing antitumor responses are thus necessary in order to expand the reach of these treatments. The inhibitor of apoptosis (IAP) protein family comprises a diverse group of proteins, many of which have immunoregulatory roles. Small molecule IAP antagonists have been developed and are undergoing early phase clinical testing. These drugs were initially developed to promote tumor cell apoptosis; however, a considerable body of work now indicates that IAP antagonists induce antitumor activity through modulation of innate and adaptive immunity. Primarily through inhibition of cellular (c)-IAP1 and c-IAP2, IAP antagonists can activate alternative NF-κB signaling, promoting B-cell survival, activation of dendritic cells and delivering a broad co-stimulatory signal to T cells. At the same time, IAP antagonists can promote tumor cell intrinsic sensitization to innate immune signals, and enhance tumor cell killing by inflammatory cytokines and phagocytic macrophages. These drugs thus represent an attractive investigational approach to immunotherapy, providing a positive signaling counterpart to the relief of signal inhibition conferred by checkpoint blockade.
Collapse
Affiliation(s)
- Stephanie K Dougan
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Michael Dougan
- Harvard Medical School, Boston, MA 02115, USA.,Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
21
|
Schmidt N, Haydn T, Schneider I, Busch H, Boerries M, Fulda S. Smac mimetic induces an early wave of gene expression via NF-κB and AP-1 and a second wave via TNFR1 signaling. Cancer Lett 2018; 421:170-185. [DOI: 10.1016/j.canlet.2018.01.082] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 01/07/2023]
|
22
|
Romagny S, Bouaouiche S, Lucchi G, Ducoroy P, Bertoldo JB, Terenzi H, Bettaieb A, Plenchette S. S-Nitrosylation of cIAP1 Switches Cancer Cell Fate from TNFα/TNFR1-Mediated Cell Survival to Cell Death. Cancer Res 2018; 78:1948-1957. [PMID: 29431638 DOI: 10.1158/0008-5472.can-17-2078] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/21/2017] [Accepted: 01/31/2018] [Indexed: 11/16/2022]
Abstract
TNFα is a prominent proinflammatory cytokine and a critical mediator for the development of many types of cancer such as breast, colon, prostate, cervical, skin, liver, and chronic lymphocytic leukemia. Binding of TNFα to TNFR1 can lead to divergent signaling pathways promoting predominantly NF-κB activation but also cell death. We report here that the nitric oxide (NO) donor glyceryl trinitrate (GTN) converts TNFα, generated from immune cells or cancer cells stimulated by chemotherapy, into a prodeath mediator in colon and mammary cancer cells. GTN-mediated S-nitrosylation of cIAP1 on cysteines 571 and 574 inhibited its E3 ubiquitin ligase activity, which in turn reduced Lys63-linked ubiquitination of RIP1 and initiated assembly of a death complex. These findings provide insights into how NO can harness advantageous aspects of inflammation in cancer and provide new therapeutic strategies.Significance: Combination of an NO donor with chemotherapeutic drug-induced TNFα represents a potentially valuable anticancer strategy. Cancer Res; 78(8); 1948-57. ©2018 AACR.
Collapse
Affiliation(s)
- Sabrina Romagny
- EPHE, PSL Research University, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche-Comté, Dijon, France
| | - Sarra Bouaouiche
- EPHE, PSL Research University, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche-Comté, Dijon, France
| | - Géraldine Lucchi
- Université de Bourgogne Franche-Comté, Clinical and Innovation Proteomic Platform, Dijon, France
| | - Patrick Ducoroy
- Université de Bourgogne Franche-Comté, Clinical and Innovation Proteomic Platform, Dijon, France
| | - Jean B Bertoldo
- Centro de Biologia Molecular Estrutural-Universidade Federal de Santa Catarina, Campus Trindade, Florianópolis, Santa Catarina, Brasil
| | - Hernan Terenzi
- Centro de Biologia Molecular Estrutural-Universidade Federal de Santa Catarina, Campus Trindade, Florianópolis, Santa Catarina, Brasil
| | - Ali Bettaieb
- EPHE, PSL Research University, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche-Comté, Dijon, France
| | - Stéphanie Plenchette
- EPHE, PSL Research University, Paris, France. .,LIIC, EA7269, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
23
|
Ros U, Peña-Blanco A, Hänggi K, Kunzendorf U, Krautwald S, Wong WWL, García-Sáez AJ. Necroptosis Execution Is Mediated by Plasma Membrane Nanopores Independent of Calcium. Cell Rep 2017; 19:175-187. [PMID: 28380356 PMCID: PMC5465952 DOI: 10.1016/j.celrep.2017.03.024] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/22/2016] [Accepted: 03/06/2017] [Indexed: 01/14/2023] Open
Abstract
Necroptosis is a form of regulated necrosis that
results in cell death and content release after plasma membrane permeabilization.
However, little is known about the molecular events responsible for the disruption of
the plasma membrane. Here, we find that early increase in cytosolic calcium in
TNF-induced necroptosis is mediated by treatment with a Smac mimetic via the
TNF/RIP1/TAK1 survival pathway. This does not require the activation of the necrosome
and is dispensable for necroptosis. Necroptosis induced by the activation of TLR3/4
pathways does not trigger early calcium flux. We also demonstrate that necroptotic
plasma membrane rupture is mediated by osmotic forces and membrane pores around 4 nm
in diameter. This late permeabilization step represents a hallmark in necroptosis
execution that is cell and treatment independent and requires the RIP1/RIP3/MLKL
core. In support of this, treatment with osmoprotectants reduces cell damage in an
in vivo necroptosis model of ischemia-reperfusion injury. Early calcium signaling in TSZ necroptosis correlates with
cellular levels of cIAP1/2 Calcium flux is induced by a Smac mimetic and is
dispensable for necroptosis execution Pores ∼4 nm in diameter mediate final plasma membrane
disruption in necroptosis
Collapse
Affiliation(s)
- Uris Ros
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany.
| | - Aida Peña-Blanco
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany
| | - Kay Hänggi
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany; Max-Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany.
| |
Collapse
|
24
|
Modulation of apoptotic response by LAR family phosphatases-cIAP1 signaling during urinary tract morphogenesis. Proc Natl Acad Sci U S A 2017; 114:E9016-E9025. [PMID: 29073098 DOI: 10.1073/pnas.1707229114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The elimination of unwanted cells by apoptosis is necessary for tissue morphogenesis. However, the cellular control of morphogenetic apoptosis is poorly understood, notably the modulation of cell sensitivity to apoptotic stimuli. Ureter maturation, the process by which the ureter is displaced to the bladder wall, represents an exquisite example of morphogenetic apoptosis, requiring the receptor protein tyrosine phosphatases (RPTPs): LAR and RPTPσ. Here we show that LAR-RPTPs act through cellular inhibitor of apoptosis protein 1 (cIAP1) to modulate caspase 3,7-mediated ureter maturation. Pharmacologic or genetic inactivation of cIAP1 reverts the apoptotic deficit of LAR-RPTP-deficient embryos. Moreover, Birc2 (cIAP1) inactivation generates excessive apoptosis leading to vesicoureteral reflux in newborns, which underscores the importance of apoptotic modulation during urinary tract morphogenesis. We finally demonstrate that LAR-RPTP deficiency increases cIAP1 stability during apoptotic cell death. Together these results identify a mode of cIAP1 regulation playing a critical role in the cellular response to apoptotic pathway activation in the embryo.
Collapse
|
25
|
Abstract
Inhibitor of apoptosis proteins (IAPs) are critical regulators of cell death and survival pathways. Mice lacking cIAP1 and either cIAP2 or XIAP die in utero, and myeloid lineage-specific deletion of all IAPs causes sterile inflammation, but their role in the skin is unknown. We generated epidermal-specific IAP-deficient mice and found that combined genetic deletion of cIAP1 (epidermal knockout [EKO]) in keratinocytes and ubiquitous cIAP2 deletion (cIap1EKO/EKO.cIap2-/-) caused profound skin inflammation and keratinocyte death, lethal by postpartum day 10. To investigate their role in skin homeostasis, we injected an IAP antagonist compound subcutaneously into wild-type and knockout mice. This induced a toxic epidermal necrolysis-like local inflammation, which mirrored the phenotype seen in cIap1EKO/EKO.cIap2-/- mice. Loss of one Ripk1 allele limited lesion formation and significantly extended the lifespan of cIap1EKO/EKO.cIap2-/- mice. cIAP activities are important for recruitment of LUBAC to signaling complexes, and loss of LUBAC component SHARPIN, induces dermatitis in mice. Consistent with this relationship between cIAPs and LUBAC, Ripk1 heterozygosity also protected against development of dermatitis in Sharpin-deficient mice. This work therefore refines our molecular understanding of inflammatory signaling in the skin and defines potential targets for treating skin inflammation.
Collapse
|
26
|
Pilling AB, Hwang O, Boudreault A, Laurent A, Hwang C. IAP Antagonists Enhance Apoptotic Response to Enzalutamide in Castration-Resistant Prostate Cancer Cells via Autocrine TNF-α Signaling. Prostate 2017; 77:866-877. [PMID: 28240376 DOI: 10.1002/pros.23327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/03/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) remains incurable and identifying effective treatments continues to present a clinical challenge. Although treatment with enzalutamide, a second generation androgen receptor (AR) antagonist, prolongs survival in prostate cancer patients, responses can be limited by intrinsic resistance or acquired resistance. A potential mechanism of resistance to androgen axis inhibition is evasion of apoptosis. Inhibitor of apoptosis proteins (IAPs) are found to be overexpressed in prostate cancer and function to block apoptosis and promote survival signaling. Novel, small-molecule IAP antagonists, such as AEG40995, are emerging as a strategy to induce apoptosis and increase therapeutic response in cancer. METHODS Human prostate cancer cell lines LNCaP and C4-2 were treated with enzalutamide with or without addition of IAP antagonist AEG40995 and proliferation and survival were determined by MTS and clonogenic assay. Western blot was used to evaluate IAP protein expression changes and PARP-1 cleavage was assessed as indication of apoptosis. Flow cytometry was performed to analyze apoptosis in treated cells. Caspase activity was determined by luminescence assay. Quantitative real-time PCR and immunometric ELISA was used to assess TNF-α (transcript and protein levels, respectively) in response to treatment. RESULTS In this study, we demonstrate that IAP antagonist AEG40995 exhibits minimal effects on prostate cancer cell proliferation or survival, but rapidly degrades cIAP1 protein. Combination treatment with enzalutamide demonstrates that AEG40995 increases apoptosis and reduces proliferation and clonogenic survival in cell line models of prostate cancer. Mechanistically, we demonstrate that apoptosis in response to enzalutamide and IAP antagonist requires activation of caspase-8, suggesting extrinsic/death receptor apoptosis signaling. Assessment of TNF-α in response to combination treatment with enzalutamide and AEG40995 reveals increased mRNA expression and autocrine protein secretion. Blocking TNF-α signaling abrogates the apoptotic response demonstrating that TNF-α plays a critical role in executing cell death in response to this drug combination. CONCLUSIONS These findings suggest that IAP antagonists can increase sensitivity and amplify the caspase-mediated apoptotic response to enzalutamide through TNF-α signaling mechanisms. Combination with an IAP antagonist increases enzalutamide sensitivity, lowers the apoptotic threshold and may combat drug resistance in patients with prostate cancer. Prostate 77:866-877, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amanda B Pilling
- Henry Ford Health System, Henry Ford Cancer Institute, Detroit, Michigan
| | - Ok Hwang
- Henry Ford Health System, Henry Ford Cancer Institute, Detroit, Michigan
| | | | | | - Clara Hwang
- Henry Ford Health System, Henry Ford Cancer Institute, Detroit, Michigan
| |
Collapse
|
27
|
Vasilikos L, Spilgies LM, Knop J, Wong WWL. Regulating the balance between necroptosis, apoptosis and inflammation by inhibitors of apoptosis proteins. Immunol Cell Biol 2017; 95:160-165. [PMID: 27904150 DOI: 10.1038/icb.2016.118] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 02/06/2023]
Abstract
Understanding how inhibitor of apoptosis proteins (IAPs) regulate apoptosis and necroptosis has been fast-forwarded by the use of Smac mimetics (SMs) to deplete or inhibit the IAPs, specifically cIAP1, cIAP2 and XIAP. The loss or inhibition of cIAP1, cIAP2 and XIAP causes the majority of cells to be sensitized to death receptor induced cell death, such as with tumour necrosis factor (TNF). Mouse genetics shows that there is some functional redundancy and the use of SMs has allowed us to understand how changing the composition of proteins recruited to TNF receptor 1 on TNF ligation can alter protein complex formation and activation of apoptosis or necroptosis, particularly when caspases are inhibited. Determining when or how caspase inhibition occurs physiologically combined with the loss of IAPs will be the next challenge in understanding the ability of IAPs to prevent cell death and/or limit inflammation.
Collapse
Affiliation(s)
- Lazaros Vasilikos
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Lisanne M Spilgies
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Janin Knop
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Wendy Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Zappavigna S, Scuotto M, Cossu AM, Ingrosso D, De Rosa M, Schiraldi C, Filosa R, Caraglia M. The 1,4 benzoquinone-featured 5-lipoxygenase inhibitor RF-Id induces apoptotic death through downregulation of IAPs in human glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:167. [PMID: 27770821 PMCID: PMC5075202 DOI: 10.1186/s13046-016-0440-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/06/2016] [Indexed: 12/23/2022]
Abstract
Background Embelin is a potent dual inhibitor of 5-lipoxigenase (5-LOX) and microsomal prostaglandin E2 synthase (mPGES)-1 that suppresses proliferation of human glioma cells and induces apoptosis by inhibiting XIAP and NF-κB signaling pathway. Synthetic structural modification yielded the derivative 3-((decahydronaphthalen-6-yl)methyl)-2,5-dihydroxycyclohexa-2,5-diene-1,4-dione (RF-Id), an embelin constrained analogue, with improved efficiency against 5-LOX in human neutrophils and anti-inflammatory activity in vivo. Taking into account that lipoxygenase (LOX) metabolites, from arachidonic acid and linoleic acid, have been implicated in tumor progression, here, we determined whether RF-Id was able to hinder glioblastoma (GBM) cancer cell growth and the related mechanisms. Methods U87MG and LN229 cells were plated in 96-wells and treated with increasing concentrations of RF-Id. Cell viability was evaluated by MTT assay. The effects of the compounds on cell cycle, apoptosis, oxidative stress and autophagy were assessed by flow cytometry (FACS). The mode of action was confirmed by Taqman apoptosis array and evaluating caspase cascade and NFκB pathway by western blotting technique. Results Here, we found that RF-Id induced a stronger inhibition of GBM cell growth than treatment with embelin. Flow cytometry analysis showed that RF-Id induced about 30 % apoptosis and a slight increase of autophagy after 72 h on U87-MG cells. Moreover, the compound induced an increase in the percentage of cells in G2 and S phase that was paralleled by an increase of p21 and p27 expression but no significant changes of the mitochondrial membrane potential; array analysis showed a significant upregulation of CASP8 and a downregulation of IAP family and NFκB genes in cells treated with RF-Id. RF-Id induced a significant cleavage of caspases 8, 9, 3 and 7, blocked c-IAP2/XIAP interaction by inducing XIAP degradation and inhibited NFκB pathway. Conclusions RF-Id induced a caspase-dependent apoptosis in GBM cells by inhibiting IAP family proteins and NFκB pathway and represents a promising lead compound for designing a new class of anti-cancer drugs with multiple targets. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0440-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- S Zappavigna
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. De Crecchio 7, Naples, 80138, Italy
| | - M Scuotto
- Department of Experimental Medicine, Second University of Naples, Via L. De Crecchio, 7, Naples, 80138, Italy
| | - A M Cossu
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. De Crecchio 7, Naples, 80138, Italy
| | - D Ingrosso
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. De Crecchio 7, Naples, 80138, Italy
| | - M De Rosa
- Department of Experimental Medicine, Second University of Naples, Via L. De Crecchio, 7, Naples, 80138, Italy
| | - C Schiraldi
- Department of Experimental Medicine, Second University of Naples, Via L. De Crecchio, 7, Naples, 80138, Italy
| | - R Filosa
- Department of Experimental Medicine, Second University of Naples, Via L. De Crecchio, 7, Naples, 80138, Italy.
| | - M Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, via L. De Crecchio 7, Naples, 80138, Italy.
| |
Collapse
|
29
|
Peltzer N, Darding M, Walczak H. Holding RIPK1 on the Ubiquitin Leash in TNFR1 Signaling. Trends Cell Biol 2016; 26:445-461. [DOI: 10.1016/j.tcb.2016.01.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/22/2022]
|
30
|
Dondelinger Y, Darding M, Bertrand MJM, Walczak H. Poly-ubiquitination in TNFR1-mediated necroptosis. Cell Mol Life Sci 2016; 73:2165-76. [PMID: 27066894 PMCID: PMC4887548 DOI: 10.1007/s00018-016-2191-4] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/28/2022]
Abstract
Tumor necrosis factor (TNF) is a master pro-inflammatory cytokine, and inappropriate TNF signaling is implicated in the pathology of many inflammatory diseases. Ligation of TNF to its receptor TNFR1 induces the transient formation of a primary membrane-bound signaling complex, known as complex I, that drives expression of pro-survival genes. Defective complex I activation results in induction of cell death, in the form of apoptosis or necroptosis. This switch occurs via internalization of complex I components and assembly and activation of secondary cytoplasmic death complexes, respectively known as complex II and necrosome. In this review, we discuss the crucial regulatory functions of ubiquitination—a post-translational protein modification consisting of the covalent attachment of ubiquitin, and multiples thereof, to target proteins—to the various steps of TNFR1 signaling leading to necroptosis.
Collapse
Affiliation(s)
- Yves Dondelinger
- Inflammation Research Center, VIB, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium
| | - Maurice Darding
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Mathieu J M Bertrand
- Inflammation Research Center, VIB, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium.
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
31
|
Cildir G, Low KC, Tergaonkar V. Noncanonical NF-κB Signaling in Health and Disease. Trends Mol Med 2016; 22:414-429. [PMID: 27068135 DOI: 10.1016/j.molmed.2016.03.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/18/2022]
Abstract
Noncanonical NF-κB signaling differs from canonical NF-κB signaling by being activated through different cell surface receptors, cytoplasmic adaptors, and NF-κB dimers. Under normal physiological conditions, this noncanonical pathway has been implicated in diverse biological processes, including lymphoid organogenesis, B cell maturation, osteoclast differentiation, and various functions of other immune cells. Recently, dysfunction of this pathway has also been causally associated with numerous immune-mediated pathologies and human malignancies. Here, we summarize the core elements as well as the recently identified novel regulators of the noncanonical NF-κB signaling pathway. The involvement of this pathway in different pathologies and the potential therapeutic options that are currently envisaged are also discussed.
Collapse
Affiliation(s)
- Gökhan Cildir
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Kee Chung Low
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore; Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia.
| |
Collapse
|
32
|
Moulin M, Voss AK, Thomas T, Wong WWL, Cook WD, Koentgen F, Vince J, Silke J, Vaux DL. Response to Heard et al. EMBO J 2015; 34:2396-7. [PMID: 26427759 DOI: 10.15252/embj.201592761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
| | - Anne K Voss
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wendy Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Wendy D Cook
- La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Vic., Australia
| | | | - James Vince
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - John Silke
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - David L Vaux
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
33
|
Heard KN, Bertrand MJM, Barker PA. cIAP2 supports viability of mice lacking cIAP1 and XIAP. EMBO J 2015; 34:2393-5. [PMID: 26427758 DOI: 10.15252/embj.201592060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Kristen N Heard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Mathieu J M Bertrand
- VIB Inflammation Research Centre and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Philip A Barker
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
34
|
Baculovirus Inhibitor-of-Apoptosis Op-IAP3 Blocks Apoptosis by Interaction with and Stabilization of a Host Insect Cellular IAP. J Virol 2015; 90:533-44. [PMID: 26491164 DOI: 10.1128/jvi.02320-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/14/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Baculovirus-encoded inhibitor of apoptosis (IAP) proteins likely evolved from their host cell IAP homologs, which function as critical regulators of cell death. Despite their striking relatedness to cellular IAPs, including the conservation of two baculovirus IAP repeat (BIR) domains and a C-terminal RING, viral IAPs use an unresolved mechanism to suppress apoptosis in insects. To define this mechanism, we investigated Op-IAP3, the prototypical IAP from baculovirus OpMNPV. We found that Op-IAP3 forms a stable complex with SfIAP, the native, short-lived IAP of host insect Spodoptera frugiperda. Long-lived Op-IAP3 prevented virus-induced SfIAP degradation, which normally causes caspase activation and apoptosis. In uninfected cells, Op-IAP3 also increased SfIAP steady-state levels and extended SfIAP's half-life. Conversely, SfIAP stabilization was lost or reversed in the presence of mutated Op-IAP3 that was engineered for reduced stability. Thus, Op-IAP3 stabilizes SfIAP and preserves its antiapoptotic function. In contrast to SfIAP, Op-IAP3 failed to bind or inhibit native Spodoptera caspases. Furthermore, BIR mutations that abrogate binding of well-conserved IAP antagonists did not affect Op-IAP3's capacity to prevent virus-induced apoptosis. Remarkably, Op-IAP3 also failed to prevent apoptosis when endogenous SfIAP was ablated by RNA silencing. Thus, Op-IAP3 requires SfIAP as a cofactor. Our findings suggest a new model wherein Op-IAP3 interacts directly with SfIAP to maintain its intracellular level, thereby suppressing virus-induced apoptosis indirectly. Consistent with this model, Op-IAP3 has evolved an intrinsic stability that may serve to repress signal-induced turnover and autoubiquitination when bound to its targeted cellular IAP. IMPORTANCE The IAPs were first discovered in baculoviruses because of their potency for preventing apoptosis. However, the antiapoptotic mechanism of viral IAPs in host insects has been elusive. We show here that the prototypical viral IAP, Op-IAP3, blocks apoptosis indirectly by associating with unstable, autoubiquitinating host IAP in such a way that cellular IAP levels and antiapoptotic activities are maintained. This mechanism explains Op-IAP3's requirement for native cellular IAP as a cofactor and the dispensability of caspase inhibition. Viral IAP-mediated preservation of the host IAP homolog capitalizes on normal IAP-IAP interactions and is likely the result of viral IAP evolution in which degron-mediated destabilization and ubiquitination potential have been reduced. This mechanism illustrates another novel means by which DNA viruses incorporate host death regulators that are modified for resistance to host regulatory controls for the purpose of suppressing host cell apoptosis and acquiring replication advantages.
Collapse
|
35
|
Chaudhary AK, Yadav N, Bhat TA, O'Malley J, Kumar S, Chandra D. A potential role of X-linked inhibitor of apoptosis protein in mitochondrial membrane permeabilization and its implication in cancer therapy. Drug Discov Today 2015; 21:38-47. [PMID: 26232549 DOI: 10.1016/j.drudis.2015.07.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 12/17/2022]
Abstract
X-chromosome-linked inhibitor of apoptosis protein (XIAP) has an important regulatory role in programmed cell death by inhibiting the caspase cascade. Activation of XIAP-dependent signaling culminates into regulation of multiple cellular processes including apoptosis, innate immunity, epithelial-to-mesenchymal transition, cell migration, invasion, metastasis and differentiation. Although XIAP localizes to the cytosolic compartment, XIAP-mediated cellular signaling encompasses mitochondrial and post-mitochondrial levels. Recent findings demonstrate that XIAP also localizes to mitochondria and regulates mitochondria functions. XIAP acts upstream of mitochondrial cytochrome c release and modulates caspase-dependent apoptosis. The new function of XIAP has potential to enhance mitochondrial membrane permeabilization and other cellular functions controlling cytochrome c release. These findings could exploit the overexpression of XIAP in human tumors for therapeutic benefits.
Collapse
Affiliation(s)
- Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
36
|
Silke J, Vaux DL. IAP gene deletion and conditional knockout models. Semin Cell Dev Biol 2015; 39:97-105. [DOI: 10.1016/j.semcdb.2014.12.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/10/2023]
|
37
|
Choi JS, Park BC, Chi SW, Bae KH, Kim S, Cho S, Son WC, Myung PK, Kim JH, Park SG. HAX1 regulates E3 ubiquitin ligase activity of cIAPs by promoting their dimerization. Oncotarget 2014; 5:10084-99. [PMID: 25275296 PMCID: PMC4259407 DOI: 10.18632/oncotarget.2459] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/06/2014] [Indexed: 12/03/2022] Open
Abstract
HS-1-associated protein X-1 (HAX1) is a multi-functional protein which was first identified as a Hematopoietic cell specific Lyn Substrate 1 (HS1)-binding protein. Although the roles of HAX1 in apoptosis have been unraveled and HAX1 has been proposed to be involved in several diseases, additional roles of HAX1 are still being identified. Here, we demonstrated that HAX1 directly interacted with cellular Inhibitor of Apoptosis Proteins (cIAPs), ubiquitin E3 ligases which regulate the abundance of cellular proteins, via ubiquitin-dependent proteasomal degradation. We showed that HAX1 promotes auto-ubiquitination and degradation of cIAPs by facilitating the intermolecular homodimerization of RING finger domain. Moreover, HAX1 regulates the non-canonical Nuclear Factor-κB (NF-κB) signaling pathway by modulating the stability of NF-κB-Inducing Kinase (NIK), which is one of the substrates of cIAPs. Taken together, these results unveil a novel role of HAX1 in the non-canonical NF-κB pathway, and provide an important clue that HAX1 is a potential therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Jin Sun Choi
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Byoung Chul Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seung Wook Chi
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Cell Function Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sunhong Kim
- Targeted Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Woo-Chan Son
- Asan Institute for Life Sciences and Asan Medical Center, Seoul, Republic of Korea
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Pyung Keun Myung
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Hoon Kim
- Targeted Gene Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, Daejeon, Republic of Korea
| | - Sung Goo Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| |
Collapse
|
38
|
Bai L, Smith DC, Wang S. Small-molecule SMAC mimetics as new cancer therapeutics. Pharmacol Ther 2014; 144:82-95. [PMID: 24841289 PMCID: PMC4247261 DOI: 10.1016/j.pharmthera.2014.05.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/19/2022]
Abstract
Apoptosis is a tightly regulated cellular process and faulty regulation of apoptosis is a hallmark of human cancers. Targeting key apoptosis regulators with the goal to restore apoptosis in tumor cells has been pursued as a new cancer therapeutic strategy. XIAP, cIAP1, and cIAP2, members of inhibitor of apoptosis (IAP) proteins, are critical regulators of cell death and survival and are attractive targets for new cancer therapy. The SMAC/DIABLO protein is an endogenous antagonist of XIAP, cIAP1, and cIAP2. In the last decade, intense research efforts have resulted in the design and development of several small-molecule SMAC mimetics now in clinical trials for cancer treatment. In this review, we will discuss the roles of XIAP, cIAP1, and cIAP2 in regulation of cell death and survival, and the design and development of small-molecule SMAC mimetics as novel cancer treatments.
Collapse
Affiliation(s)
- Longchuan Bai
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - David C Smith
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| | - Shaomeng Wang
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
39
|
Condon SM, Mitsuuchi Y, Deng Y, LaPorte MG, Rippin SR, Haimowitz T, Alexander MD, Kumar PT, Hendi MS, Lee YH, Benetatos CA, Yu G, Kapoor GS, Neiman E, Seipel ME, Burns JM, Graham MA, McKinlay MA, Li X, Wang J, Shi Y, Feltham R, Bettjeman B, Cumming MH, Vince JE, Khan N, Silke J, Day CL, Chunduru SK. Birinapant, a smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies. J Med Chem 2014; 57:3666-77. [PMID: 24684347 DOI: 10.1021/jm500176w] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Birinapant (1) is a second-generation bivalent antagonist of IAP proteins that is currently undergoing clinical development for the treatment of cancer. Using a range of assays that evaluated cIAP1 stability and oligomeric state, we demonstrated that 1 stabilized the cIAP1-BUCR (BIR3-UBA-CARD-RING) dimer and promoted autoubiquitylation of cIAP1 in vitro. Smac-mimetic 1-induced loss of cIAPs correlated with inhibition of TNF-mediated NF-κB activation, caspase activation, and tumor cell killing. Many first-generation Smac-mimetics such as compound A (2) were poorly tolerated. Notably, animals that lack functional cIAP1, cIAP2, and XIAP are not viable, and 2 mimicked features of triple IAP knockout cells in vitro. The improved tolerability of 1 was associated with (i) decreased potency against cIAP2 and affinity for XIAP BIR3 and (ii) decreased ability to inhibit XIAP-dependent signaling pathways. The P2' position of 1 was critical to this differential activity, and this improved tolerability has allowed 1 to proceed into clinical studies.
Collapse
Affiliation(s)
- Stephen M Condon
- TetraLogic Pharmaceuticals, Inc. , 343 Phoenixville Pike, Malvern, Pennsylvania 19355, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Guicciardi ME, Werneburg NW, Bronk SF, Franke A, Yagita H, Thomas G, Gores GJ. Cellular inhibitor of apoptosis (cIAP)-mediated ubiquitination of phosphofurin acidic cluster sorting protein 2 (PACS-2) negatively regulates tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) cytotoxicity. PLoS One 2014; 9:e92124. [PMID: 24633224 PMCID: PMC3954888 DOI: 10.1371/journal.pone.0092124] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/18/2014] [Indexed: 01/24/2023] Open
Abstract
Lysosomal membrane permeabilization is an essential step in TRAIL-induced apoptosis of liver cancer cell lines. TRAIL-induced lysosomal membrane permeabilization is mediated by the multifunctional sorting protein PACS-2 and repressed by the E3 ligases cIAP-1 and cIAP-2. Despite the opposing roles for PACS-2 and cIAPs in TRAIL-induced apoptosis, an interaction between these proteins has yet to be examined. Herein, we report that cIAP-1 and cIAP-2 confer TRAIL resistance to hepatobiliary cancer cell lines by reducing PACS-2 levels. Under basal conditions, PACS-2 underwent K48-linked poly-ubiquitination, resulting in PACS-2 proteasomal degradation. Biochemical assays showed cIAP-1 and cIAP-2 interacted with PACS-2 in vitro and co-immunoprecipitation studies demonstrated that the two cIAPs bound PACS-2 in vivo. More importantly, both cIAP-1 and cIAP-2 directly mediated PACS-2 ubiquitination in a cell-free assay. Single c-Iap-1 or c-Iap-2 gene knock-outs in mouse hepatocytes did not lead to PACS-2 accumulation. However, deletion of both cIAP-1 and cIAP-2 reduced PACS-2 ubiquitination, which increased PACS-2 levels and sensitized HuH-7 cells to TRAIL-induced lysosomal membrane permeabilization and apoptosis. Correspondingly, deletion of cIAPs sensitized wild-type, but not PACS-2-deficient hepatocarcinoma cells or Pacs-2−/− mouse hepatocytes to TRAIL-induced apoptosis. Together, these data suggest cIAPs constitutively downregulate PACS-2 by polyubiquitination and proteasomal degradation, thereby restraining TRAIL-induced killing of liver cancer cells.
Collapse
Affiliation(s)
- Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Nathan W. Werneburg
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Steven F. Bronk
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Adrian Franke
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, Tokyo, Japan
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
41
|
Guo S, Messmer-Blust AF, Wu J, Song X, Philbrick MJ, Shie JL, Rana JS, Li J. Role of A20 in cIAP-2 protection against tumor necrosis factor α (TNF-α)-mediated apoptosis in endothelial cells. Int J Mol Sci 2014; 15:3816-33. [PMID: 24595242 PMCID: PMC3975369 DOI: 10.3390/ijms15033816] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/30/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor α (TNF-α) influences endothelial cell viability by altering the regulatory molecules involved in induction or suppression of apoptosis. However, the underlying mechanisms are still not completely understood. In this study, we demonstrated that A20 (also known as TNFAIP3, tumor necrosis factor α-induced protein 3, and an anti-apoptotic protein) regulates the inhibitor of apoptosis protein-2 (cIAP-2) expression upon TNF-α induction in endothelial cells. Inhibition of A20 expression by its siRNA resulted in attenuating expression of TNF-α-induced cIAP-2, yet not cIAP-1 or XIAP. A20-induced cIAP-2 expression can be blocked by the inhibition of phosphatidyl inositol-3 kinase (PI3-K), but not nuclear factor (NF)-κB, while concomitantly increasing the number of endothelial apoptotic cells and caspase 3 activation. Moreover, TNF-α-mediated induction of apoptosis was enhanced by A20 inhibition, which could be rescued by cIAP-2. Taken together, these results identify A20 as a cytoprotective factor involved in cIAP-2 inhibitory pathway of TNF-α-induced apoptosis. This is consistent with the idea that endothelial cell viability is dependent on interactions between inducers and suppressors of apoptosis, susceptible to modulation by TNF-α.
Collapse
Affiliation(s)
- Shuzhen Guo
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Angela F Messmer-Blust
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jiaping Wu
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Xiaoxiao Song
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Melissa J Philbrick
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jue-Lon Shie
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jamal S Rana
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jian Li
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
42
|
Benetatos CA, Mitsuuchi Y, Burns JM, Neiman EM, Condon SM, Yu G, Seipel ME, Kapoor GS, Laporte MG, Rippin SR, Deng Y, Hendi MS, Tirunahari PK, Lee YH, Haimowitz T, Alexander MD, Graham MA, Weng D, Shi Y, McKinlay MA, Chunduru SK. Birinapant (TL32711), a bivalent SMAC mimetic, targets TRAF2-associated cIAPs, abrogates TNF-induced NF-κB activation, and is active in patient-derived xenograft models. Mol Cancer Ther 2014; 13:867-79. [PMID: 24563541 DOI: 10.1158/1535-7163.mct-13-0798] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The acquisition of apoptosis resistance is a fundamental event in cancer development. Among the mechanisms used by cancer cells to evade apoptosis is the dysregulation of inhibitor of apoptosis (IAP) proteins. The activity of the IAPs is regulated by endogenous IAP antagonists such as SMAC (also termed DIABLO). Antagonism of IAP proteins by SMAC occurs via binding of the N-terminal tetrapeptide (AVPI) of SMAC to selected BIR domains of the IAPs. Small molecule compounds that mimic the AVPI motif of SMAC have been designed to overcome IAP-mediated apoptosis resistance of cancer cells. Here, we report the preclinical characterization of birinapant (TL32711), a bivalent SMAC-mimetic compound currently in clinical trials for the treatment of cancer. Birinapant bound to the BIR3 domains of cIAP1, cIAP2, XIAP, and the BIR domain of ML-IAP in vitro and induced the autoubiquitylation and proteasomal degradation of cIAP1 and cIAP2 in intact cells, which resulted in formation of a RIPK1:caspase-8 complex, caspase-8 activation, and induction of tumor cell death. Birinapant preferentially targeted the TRAF2-associated cIAP1 and cIAP2 with subsequent inhibition of TNF-induced NF-κB activation. The activity of a variety of chemotherapeutic cancer drugs was potentiated by birinapant both in a TNF-dependent or TNF-independent manner. Tumor growth in multiple primary patient-derived xenotransplant models was inhibited by birinapant at well-tolerated doses. These results support the therapeutic combination of birinapant with multiple chemotherapies, in particular, those therapies that can induce TNF secretion.
Collapse
Affiliation(s)
- Christopher A Benetatos
- Authors' Affiliations: TetraLogic Pharmaceuticals, 343 Phoenixville Pike, Malvern, Pennsylvania; and Tsinghua University School of Medicine, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Enwere EK, Lacasse EC, Adam NJ, Korneluk RG. Role of the TWEAK-Fn14-cIAP1-NF-κB Signaling Axis in the Regulation of Myogenesis and Muscle Homeostasis. Front Immunol 2014; 5:34. [PMID: 24550918 PMCID: PMC3913901 DOI: 10.3389/fimmu.2014.00034] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/21/2014] [Indexed: 12/16/2022] Open
Abstract
Mammalian skeletal muscle maintains a robust regenerative capacity throughout life, largely due to the presence of a stem cell population known as “satellite cells” in the muscle milieu. In normal conditions, these cells remain quiescent; they are activated upon injury to become myoblasts, which proliferate extensively and eventually differentiate and fuse to form new multinucleated muscle fibers. Recent findings have identified some of the factors, including the cytokine TNFα-like weak inducer of apoptosis (TWEAK), which govern these cells’ decisions to proliferate, differentiate, or fuse. In this review, we will address the functions of TWEAK, its receptor Fn14, and the associated signal transduction molecule, the cellular inhibitor of apoptosis 1 (cIAP1), in the regulation of myogenesis. TWEAK signaling can activate the canonical NF-κB signaling pathway, which promotes myoblast proliferation and inhibits myogenesis. In addition, TWEAK activates the non-canonical NF-κB pathway, which, in contrast, promotes myogenesis by increasing myoblast fusion. Both pathways are regulated by cIAP1, which is an essential component of downstream signaling mediated by TWEAK and similar cytokines. This review will focus on the seemingly contradictory roles played by TWEAK during muscle regeneration, by highlighting the interplay between the two NF-κB pathways under physiological and pathological conditions. We will also discuss how myogenesis is negatively affected by chronic conditions, which affect homeostasis of the skeletal muscle environment.
Collapse
Affiliation(s)
- Emeka K Enwere
- Department of Medical Microbiology and Immunology, University of Alberta , Edmonton, AB , Canada
| | - Eric C Lacasse
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada
| | - Nadine J Adam
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| | - Robert G Korneluk
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
44
|
Apoptosis in schistosomes: toward novel targets for the treatment of schistosomiasis. Trends Parasitol 2014; 30:75-84. [DOI: 10.1016/j.pt.2013.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/29/2013] [Accepted: 12/06/2013] [Indexed: 12/25/2022]
|
45
|
Yang W, Cooke M, Duckett CS, Yang X, Dorsey JF. Distinctive effects of the cellular inhibitor of apoptosis protein c-IAP2 through stabilization by XIAP in glioblastoma multiforme cells. Cell Cycle 2014; 13:992-1005. [PMID: 24552816 DOI: 10.4161/cc.27880] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inhibitor of apoptosis proteins (IAPs) are extensively involved in NFκB signaling pathways. Regulation of c-IAP2 turnover by other proteins was investigated in glioblastoma multiforme (GBM) cells in the present study. When overexpressed, X-linked IAP (XIAP) enhanced expression of ectopic c-IAP2, but not c-IAP1, and endogenous c-IAP2 levels were reduced once XIAP expression was silenced. TNFα stimulation substantially increased c-IAP2 expression, and this upregulation was impaired by suppression of XIAP. Similarly, when XIAP was limiting due to severe hypoxic conditions, c-IAP2 levels were downregulated. These data together indicate that XIAP is an important regulator responsible for stabilization of c-IAP2 levels under different conditions. Protein interactions occur through binding of BIR2 and BIR3 domains of c-IAP2 with the RING finger of XIAP. XIAP inhibition of c-IAP2 auto-degradation was dependent on this physical interaction, and it was independent of XIAP E3 ligase activity. Global c-IAP2 ubiquitination was not affected by XIAP, although c-IAP2 levels were significantly increased. A CARD-RING-containing fragment of c-IAP2 was found to target XIAP for proteasome-independent degradation, but it was unable to sensitize GBM cells to chemo-reagents. The XIAP-stabilized c-IAP2 was found to enhance IκB-α phosphorylation on serines 32 and 36, and to antagonize XIAP-induced increase in mature Smac and Bcl10. Taken together, our data identify a distinctive role of c-IAP2 as stabilizer of XIAP, which is likely involved in regulation of NFκB activation and apoptosis in GBM cells.
Collapse
Affiliation(s)
- Wensheng Yang
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Mariana Cooke
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Colin S Duckett
- Departments of Pathology and Internal Medicine and Comprehensive Cancer Center; University of Michigan; Ann Arbor, MI USA
| | - Xiaolu Yang
- Department of Cancer Biology and the Abramson Family Cancer Research Institute; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Jay F Dorsey
- Department of Radiation Oncology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|
46
|
Abstract
Inhibitor of apoptosis (IAP) proteins interface with, and regulate a large number of, cell signaling pathways. If there is a common theme to these pathways, it is that they are involved in the development of the immune system, immune responses, and unsurprisingly, given their name, cell death. Beyond that it is difficult to discover an underlying logic because sometimes IAPs are required to inhibit or prevent signaling, whereas in other cases they are required for signaling to take place. In whatever role they play, they are recruited into signaling complexes and function as ubiquitin E3 ligases, via their RING domains. This review discusses IAP regulation of signaling pathways and focuses on the mammalian IAPs, XIAP, c-IAP1, and c-IAP2, with a particular emphasis on techniques and methods that were used to uncover their roles. We also provide a perspective on targeting IAP proteins for therapeutic intervention and methods used to define the clinical relevance of IAP proteins.
Collapse
Affiliation(s)
- John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, California, USA.
| |
Collapse
|
47
|
Xia P, Qi Y. Cellular inhibitor of apoptosis protein-1 and survival of beta cells undergoing endoplasmic reticulum stress. VITAMINS AND HORMONES 2014; 95:269-98. [PMID: 24559922 DOI: 10.1016/b978-0-12-800174-5.00011-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pancreatic beta cells rely heavily on the endoplasmic reticulum (ER) to process folding and posttranslational modification of a large amount of insulin and many other proteins and are therefore vulnerable to ER stress. The role of the ER is thus crucial in the regulation of beta cell function and survival through the unfolded protein response (UPR) pathways. However, the UPR can either allow cells to survive by adapting to stress or kill cells through apoptosis in a context-dependent manner. How cell fate is determined following UPR activation remains enigmatic. In this review, we discuss the molecular mechanisms linking ER stress to beta cell survival or apoptosis. Specifically, we focus on the role of the cellular inhibitor of apoptosis protein-1 and propose a new model for understanding survival of beta cells undergoing ER stress.
Collapse
Affiliation(s)
- Pu Xia
- Signal Transduction Program, Centenary Institute, Sydney, Australia; Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Yanfei Qi
- Signal Transduction Program, Centenary Institute, Sydney, Australia
| |
Collapse
|
48
|
Abstract
The receptor-interacting protein kinase 3 (RIP3/RIPK3) has emerged as a critical regulator of programmed necrosis/necroptosis, an inflammatory form of cell death with important functions in pathogen-induced and sterile inflammation. RIP3 activation is tightly regulated by phosphorylation, ubiquitination, and caspase-mediated cleavage. These post-translational modifications coordinately regulate the assembly of a macromolecular signaling complex termed the necrosome. Recently, several reports indicate that RIP3 can promote inflammation independent of its pronecrotic activity. Here, we review our current understanding of the mechanisms that drive RIP3-dependent necrosis and its role in different inflammatory diseases.
Collapse
Affiliation(s)
- Kenta Moriwaki
- Department of Pathology, Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
49
|
Abstract
RIP1 kinase, a multifunctional protein that contains an N-terminal Ser/Thr kinase and a C-terminal death domain, has emerged as a key regulatory molecule involved in regulating both cell death and cell survival. When the proinflammatory cytokine TNFα stimulates its receptor, TNFR1, RIP1 regulates whether the cell lives by activating NF-κB or dies by apoptosis or necroptosis, two distinct pathways of programmed cell death that may be activated to eliminate unwanted cells. The kinase domain of RIP1 is involved in regulating necroptosis, and the death domain regulates RIP1 recruitment to the intracellular domain of TNFR1. The intermediate domain of RIP1 activates NF-κB and also interacts with RIP3 kinase, a downstream mediator of RIP1 in the execution of necroptosis. This review focuses on the functional roles of RIP1 in regulating multiple cellular mechanisms, the dynamic regulation of RIP1, and the physiological and pathological roles of RIP1 kinase in human health and disease.
Collapse
|
50
|
IAPs on the move: role of inhibitors of apoptosis proteins in cell migration. Cell Death Dis 2013; 4:e784. [PMID: 24008728 PMCID: PMC3789170 DOI: 10.1038/cddis.2013.311] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/09/2013] [Indexed: 01/06/2023]
Abstract
Inhibitors of Apoptosis Proteins (IAPs) are a class of highly conserved proteins predominantly known for the regulation of caspases and immune signaling. However, recent evidence suggests a crucial role for these molecules in the regulation of tumor cell shape and migration by controlling MAPK, NF-κB and Rho GTPases. IAPs directly control Rho GTPases, thus regulating cell shape and migration. For instance, XIAP and cIAP1 function as the direct E3 ubiquitin ligases of Rac1 and target it for proteasomal degradation. IAPs are differentially expressed in tumor cells and have been targeted by several cancer therapeutic drugs that are currently in clinical trials. Here, we summarize the current knowledge on the role of IAPs in the regulation of cell migration and discuss the possible implications of these observations in regulating tumor cell metastases.
Collapse
|