1
|
Liu F, Chen J, Li K, Li H, Zhu Y, Zhai Y, Lu B, Fan Y, Liu Z, Chen X, Jia X, Dong Z, Liu K. Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches. Mol Cancer 2024; 23:148. [PMID: 39048965 PMCID: PMC11270804 DOI: 10.1186/s12943-024-02046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
Collapse
Affiliation(s)
- Fangfang Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jingyu Chen
- Department of Pediatric Medicine, School of Third Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Kai Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Haochen Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiyi Zhu
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yubo Zhai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanle Fan
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Ziyue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojie Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xuechao Jia
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Zigang Dong
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Kangdong Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
2
|
Souza RF, Caetano MAF, Magalhães HIR, Castelucci P. Study of tumor necrosis factor receptor in the inflammatory bowel disease. World J Gastroenterol 2023; 29:2733-2746. [PMID: 37274062 PMCID: PMC10237104 DOI: 10.3748/wjg.v29.i18.2733] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/14/2023] [Accepted: 04/04/2023] [Indexed: 05/11/2023] Open
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD) are part of Inflammatory Bowel Diseases (IBD) and have pathophysiological processes such as bowel necrosis and enteric neurons and enteric glial cells. In addition, the main inflammatory mediator is related to the tumor necrosis factor-alpha (TNF-α). TNF-α is a me-diator of the intestinal inflammatory processes, thus being one of the main cytokines involved in the pathogenesis of IBD, however, its levels, when measured, are present in the serum of patients with IBD. In addition, TNF-α plays an important role in promoting inflammation, such as the production of interleukins (IL), for instance IL-1β and IL-6. There are two receptors for TNF as following: The tumor necrosis factor 1 receptor (TNFR1); and the tumor necrosis factor 2 receptor (TNFR2). They are involved in the pathogenesis of IBD and their receptors have been detected in IBD and their expression is correlated with disease activity. The soluble TNF form binds to the TNFR1 receptor with, and its activation results in a signaling cascade effects such as apoptosis, cell proliferation and cytokine secretion. In contrast, the transmembrane TNF form can bind both to TNFR1 and TNFR2. Recent studies have suggested that TNF-α is one of the main pro-inflammatory cytokines involved in the pathogenesis of IBD, since TNF levels are present in the serum of both patients with UC and CD. Intravenous and subcutaneous biologics targeting TNF-α have revolutionized the treatment of IBD, thus becoming the best available agents to induce and maintain IBD remission. The application of antibodies aimed at neutralizing TNF-α in patients with IBD that induce a satisfactory clinical response in up to 60% of patients, and also induced long-term maintenance of disease remission in most patients. It has been suggested that anti-TNF-α agents inactivate the pro-inflammatory cytokine TNF-α by direct neutralization, i.e., resulting in suppression of inflammation. However, anti-TNF-α antibodies perform more complex functions than a simple blockade.
Collapse
Affiliation(s)
- Roberta Figueiroa Souza
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | | | - Patricia Castelucci
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
3
|
cIAP1/TRAF2 interplay promotes tumor growth through the activation of STAT3. Oncogene 2023; 42:198-208. [PMID: 36400972 DOI: 10.1038/s41388-022-02544-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
Cellular inhibitor of apoptosis-1 (cIAP1) is a signaling regulator with oncogenic properties. It is involved in the regulation of signaling pathways controlling inflammation, cell survival, proliferation, differentiation and motility. It is recruited into membrane-receptor-associated signaling complexes thanks to the molecular adaptor TRAF2. However, the cIAP1/TRAF2 complex exists, independently of receptor engagement, in several subcellular compartments. The present work strengthens the importance of TRAF2 in the oncogenic properties of cIAP1. cIAPs-deficient mouse embryonic fibroblasts (MEFs) were transformed using the HRas-V12 oncogene. Re-expression of cIAP1 enhanced tumor growth in a nude mice xenograft model, and promoted lung tumor nodes formation. Deletion or mutation of the TRAF2-binding site completely abolished the oncogenic properties of cIAP1. Further, cIAP1 mediated the clustering of TRAF2, which was sufficient to stimulate tumor growth. Our TRAF2 interactome analysis showed that cIAP1 was critical for TRAF2 to bind to its protein partners. Thus, cIAP1 and TRAF2 would be two essential subunits of a signaling complex promoting a pro-tumoral signal. cIAP1/TRAF2 promoted the activation of the canonical NF-κB and ERK1/2 signaling pathways. NF-κB-dependent production of IL-6 triggered the activation of the JAK/STAT3 axis in an autocrine manner. Inhibition or downregulation of STAT3 specifically compromised the growth of cIAP1-restored MEFs but not that of MEFs expressing a cIAP1-mutant and treating mice with the STAT3 inhibitor niclosamide completely abrogated cIAP1/TRAF2-mediated tumor growth. Altogether, we demonstrate that cIAP1/TRAF2 binding is essential to promote tumor growth via the activation of the JAK/STAT3 signaling pathway.
Collapse
|
4
|
Skartsis N, Ferreira LMR, Tang Q. The dichotomous outcomes of TNFα signaling in CD4 + T cells. Front Immunol 2022; 13:1042622. [PMID: 36466853 PMCID: PMC9708889 DOI: 10.3389/fimmu.2022.1042622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
TNFa blocking agents were the first-in-class biologic drugs used for the treatment of autoimmune disease. Paradoxically, however, exacerbation of autoimmunity was observed in some patients. TNFa is a pleiotropic cytokine that has both proinflammatory and regulatory effects on CD4+ T cells and can influence the adaptive immune response against autoantigens. Here, we critically appraise the literature and discuss the intricacies of TNFa signaling that may explain the controversial findings of previous studies. The pleiotropism of TNFa is based in part on the existence of two biologically active forms of TNFa, soluble and membrane-bound, with different affinities for two distinct TNF receptors, TNFR1 and TNFR2, leading to activation of diverse downstream molecular pathways involved in cell fate decisions and immune function. Distinct membrane expression patterns of TNF receptors by CD4+ T cell subsets and their preferential binding of distinct forms of TNFα produced by a diverse pool of cellular sources during different stages of an immune response are important determinants of the differential outcomes of TNFa-TNF receptor signaling. Targeted manipulation of TNFa-TNF receptor signaling on select CD4+ T cell subsets may offer specific therapeutic interventions to dampen inflammation while fortifying immune regulation for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Nikolaos Skartsis
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Leonardo M. R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Gladstone University of California San Francisco (UCSF) Institute of Genome Immunology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
5
|
Crystallographic mining of ASK1 regulators to unravel the intricate PPI interfaces for the discovery of small molecule. Comput Struct Biotechnol J 2022; 20:3734-3754. [PMID: 35891784 PMCID: PMC9294202 DOI: 10.1016/j.csbj.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Protein seldom performs biological activities in isolation. Understanding the protein–protein interactions’ physical rewiring in response to pathological conditions or pathogen infection can help advance our comprehension of disease etiology, progression, and pathogenesis, which allow us to explore the alternate route to control the regulation of key target interactions, timely and effectively. Nonalcoholic steatohepatitis (NASH) is now a global public health problem exacerbated due to the lack of appropriate treatments. The most advanced anti-NASH lead compound (selonsertib) is withdrawn, though it is able to inhibit its target Apoptosis signal-regulating kinase 1 (ASK1) completely, indicating the necessity to explore alternate routes rather than complete inhibition. Understanding the interaction fingerprints of endogenous regulators at the molecular level that underpin disease formation and progression may spur the rationale of designing therapeutic strategies. Based on our analysis and thorough literature survey of the various key regulators and PTMs, the current review emphasizes PPI-based drug discovery’s relevance for NASH conditions. The lack of structural detail (interface sites) of ASK1 and its regulators makes it challenging to characterize the PPI interfaces. This review summarizes key regulators interaction fingerprinting of ASK1, which can be explored further to restore the homeostasis from its hyperactive states for therapeutics intervention against NASH.
Collapse
Key Words
- ASK1
- ASK1, Apoptosis signal-regulating kinase 1
- CFLAR, CASP8 and FADD-like apoptosis regulator
- CREG, Cellular repressor of E1A-stimulated genes
- DKK3, Dickkopf-related protein 3
- Interaction fingerprint
- NAFLD, Non-alcoholic fatty liver disease
- NASH
- NASH, Nonalcoholic steatohepatitis
- PPI, Protein-protein interaction
- PTM, Post-trancriptional modification
- PTMs
- Protein-protein interaction
- TNFAIP3, TNF Alpha Induced Protein 3
- TRAF2/6, Tumor necrosis factor receptor (TNFR)-associated factor2/6
- TRIM48, Tripartite Motif Containing 48
- TRX, Thioredoxin
- USP9X, Ubiquitin Specific Peptidase 9 X-Linked
Collapse
|
6
|
Park HB, Baek KH. E3 ligases and deubiquitinating enzymes regulating the MAPK signaling pathway in cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188736. [DOI: 10.1016/j.bbcan.2022.188736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022]
|
7
|
Bai J, Ding B, Li H. Targeting TNFR2 in Cancer: All Roads Lead to Rome. Front Immunol 2022; 13:844931. [PMID: 35251045 PMCID: PMC8891135 DOI: 10.3389/fimmu.2022.844931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
TNF receptor 2 (TNFR2) has become one of the best potential immune checkpoints that might be targeted, mainly because of its vital role in tumor microenvironments (TMEs). Overexpression of TNFR2 in some tumor cells and essential function in immunosuppressive cells, especially regulatory T cells (Tregs), makes blocking TNFR2 an excellent strategy in cancer treatment; however, there is evidence showing that activating TNFR2 can also inhibit tumor progression in vivo. In this review, we will discuss drugs that block and activate TNFR2 under clinical trials or preclinical developments up till now. Meanwhile, we summarize and explore the possible mechanisms related to them.
Collapse
Affiliation(s)
- Jingchao Bai
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bowen Ding
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
8
|
Mathien S, Tesnière C, Meloche S. Regulation of Mitogen-Activated Protein Kinase Signaling Pathways by the Ubiquitin-Proteasome System and Its Pharmacological Potential. Pharmacol Rev 2021; 73:263-296. [PMID: 34732541 DOI: 10.1124/pharmrev.120.000170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades are evolutionarily conserved signaling pathways that play essential roles in transducing extracellular environmental signals into diverse cellular responses to maintain homeostasis. These pathways are classically organized into an architecture of three sequentially acting protein kinases: a MAPK kinase kinase that phosphorylates and activates a MAPK kinase, which in turn phosphorylates and activates the effector MAPK. The activity of MAPKs is tightly regulated by phosphorylation of their activation loop, which can be modulated by positive and negative feedback mechanisms to control the amplitude and duration of the signal. The signaling outcomes of MAPK pathways are further regulated by interactions of MAPKs with scaffolding and regulatory proteins. Accumulating evidence indicates that, in addition to these mechanisms, MAPK signaling is commonly regulated by ubiquitin-proteasome system (UPS)-mediated control of the stability and abundance of MAPK pathway components. Notably, the biologic activity of some MAPKs appears to be regulated mainly at the level of protein turnover. Recent studies have started to explore the potential of targeted protein degradation as a powerful strategy to investigate the biologic functions of individual MAPK pathway components and as a new therapeutic approach to overcome resistance to current small-molecule kinase inhibitors. Here, we comprehensively review the mechanisms, physiologic importance, and pharmacological potential of UPS-mediated protein degradation in the control of MAPK signaling. SIGNIFICANCE STATEMENT: Accumulating evidence highlights the importance of targeted protein degradation by the ubiquitin-proteasome system in regulating and fine-tuning the signaling output of mitogen-activated protein kinase (MAPK) pathways. Manipulating protein levels of MAPK cascade components may provide a novel approach for the development of selective pharmacological tools and therapeutics.
Collapse
Affiliation(s)
- Simon Mathien
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| | - Chloé Tesnière
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada (S.Ma., C.T., S.Me.); and Molecular Biology Program, Faculty of Medicine (C.T., S.Me.) and Department of Pharmacology and Physiology (S.Me.), Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Brys R, Gibson K, Poljak T, Van Der Plas S, Amantini D. Discovery and development of ASK1 inhibitors. PROGRESS IN MEDICINAL CHEMISTRY 2020; 59:101-179. [PMID: 32362327 DOI: 10.1016/bs.pmch.2020.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant activation of mitogen-activated protein kinases (MAPKs) like c-Jun N-terminal kinase (JNK) and p38 is an event involved in the pathophysiology of numerous human diseases. The apoptosis signal-regulating kinase 1 (ASK1) is an upstream target that gets activated only under pathological conditions and as such is a promising target for therapeutic intervention. In the first part of this review the molecular mechanisms leading to ASK1 activation and regulation will be described as well as the evidences supporting a pathogenic role for ASK1 in human disease. In the second part, an update on drug discovery efforts towards the discovery and development of ASK1-targeting therapies will be provided.
Collapse
Affiliation(s)
| | - Karl Gibson
- Sandexis Medicinal Chemistry Ltd, Innovation House Discovery ParkSandwich, Kent, United Kingdom
| | | | | | | |
Collapse
|
10
|
Atretkhany KSN, Gogoleva VS, Drutskaya MS, Nedospasov SA. Distinct modes of TNF signaling through its two receptors in health and disease. J Leukoc Biol 2020; 107:893-905. [PMID: 32083339 DOI: 10.1002/jlb.2mr0120-510r] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022] Open
Abstract
TNF is a key proinflammatory and immunoregulatory cytokine whose deregulation is associated with the development of autoimmune diseases and other pathologies. Recent studies suggest that distinct functions of TNF may be associated with differential engagement of its two receptors: TNFR1 or TNFR2. In this review, we discuss the relative contributions of these receptors to pathogenesis of several diseases, with the focus on autoimmunity and neuroinflammation. In particular, we discuss the role of TNFRs in the development of regulatory T cells during neuroinflammation and recent findings concerning targeting TNFR2 with agonistic and antagonistic reagents in various murine models of autoimmune and neuroinflammatory disorders and cancer.
Collapse
Affiliation(s)
- Kamar-Sulu N Atretkhany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Violetta S Gogoleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia.,Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
11
|
Li LJ, Zheng JC, Kang R, Yan JQ. Targeting Trim69 alleviates high fat diet (HFD)-induced hippocampal injury in mice by inhibiting apoptosis and inflammation through ASK1 inactivation. Biochem Biophys Res Commun 2019; 515:658-664. [DOI: 10.1016/j.bbrc.2019.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
|
12
|
β-TrCP-dependent degradation of ASK1 suppresses the induction of the apoptotic response by oxidative stress. Biochim Biophys Acta Gen Subj 2018; 1862:2271-2280. [DOI: 10.1016/j.bbagen.2018.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022]
|
13
|
Martinez M, Rossetto IMU, Neto FSL, Tirapelli LF, Tirapelli DPC, Chuffa LGA, Cagnon VHA, Martinez FE. Interactions of ethanol and caffeine on apoptosis in the rat cerebellum (voluntary ethanol consumers). Cell Biol Int 2018; 42:1575-1583. [DOI: 10.1002/cbin.11054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/08/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Marcelo Martinez
- Department of Morphology and Pathology; Federal University of São Carlos; SP Brazil
| | | | | | - Luiz F. Tirapelli
- Department of Surgery and Anatomy; USP-University of São Paulo; Ribeirão Preto SP Brazil
| | | | | | - Valeria H. A. Cagnon
- Department of Anatomy; Cellular Biology, Physiology and Biophysics, UNICAMP; Campinas SP Brazil
| | | |
Collapse
|
14
|
Zhang P, Wang PX, Zhao LP, Zhang X, Ji YX, Zhang XJ, Fang C, Lu YX, Yang X, Gao MM, Zhang Y, Tian S, Zhu XY, Gong J, Ma XL, Li F, Wang Z, Huang Z, She ZG, Li H. The deubiquitinating enzyme TNFAIP3 mediates inactivation of hepatic ASK1 and ameliorates nonalcoholic steatohepatitis. Nat Med 2018; 24:84-94. [PMID: 29227477 DOI: 10.1038/nm.4453] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/06/2017] [Indexed: 02/06/2023]
Abstract
Activation of apoptosis signal-regulating kinase 1 (ASK1) in hepatocytes is a key process in the progression of nonalcoholic steatohepatitis (NASH) and a promising target for treatment of the condition. However, the mechanism underlying ASK1 activation is still unclear, and thus the endogenous regulators of this kinase remain open to be exploited as potential therapeutic targets. In screening for proteins that interact with ASK1 in the context of NASH, we identified the deubiquitinase tumor necrosis factor alpha-induced protein 3 (TNFAIP3) as a key endogenous suppressor of ASK1 activation, and we found that TNFAIP3 directly interacts with and deubiquitinates ASK1 in hepatocytes. Hepatocyte-specific ablation of Tnfaip3 exacerbated nonalcoholic fatty liver disease- and NASH-related phenotypes in mice, including glucose metabolism disorders, lipid accumulation and enhanced inflammation, in an ASK1-dependent manner. In contrast, transgenic or adeno-associated virus-mediated TNFAIP3 gene delivery in the liver in both mouse and nonhuman primate models of NASH substantially blocked the onset and progression of the disease. These results implicate TNFAIP3 as a functionally important endogenous suppressor of ASK1 hyperactivation in the pathogenesis of NASH and identify it as a potential new molecular target for NASH therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ling-Ping Zhao
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xin Zhang
- Institute of Model Animals of Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital, Wuhan, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Chun Fang
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yue-Xin Lu
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xia Yang
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Mao-Mao Gao
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan Zhang
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Song Tian
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jun Gong
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Feng Li
- Basic Medical School, Wuhan University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zan Huang
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital, Wuhan, China
| |
Collapse
|
15
|
Nishida T, Hattori K, Watanabe K. The regulatory and signaling mechanisms of the ASK family. Adv Biol Regul 2017; 66:2-22. [PMID: 28669716 DOI: 10.1016/j.jbior.2017.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 01/05/2023]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) was identified as a MAP3K that activates the JNK and p38 pathways, and subsequent studies have reported ASK2 and ASK3 as members of the ASK family. The ASK family is activated by various intrinsic and extrinsic stresses, including oxidative stress, ER stress and osmotic stress. Numerous lines of evidence have revealed that members of the ASK family are critical for signal transduction systems to control a wide range of stress responses such as cell death, differentiation and cytokine induction. In this review, we focus on the precise signaling mechanisms of the ASK family in response to diverse stressors.
Collapse
Affiliation(s)
- Takuto Nishida
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Kazuki Hattori
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| |
Collapse
|
16
|
Hikami S, Shiozaki A, Kitagawa-Juge M, Ichikawa D, Kosuga T, Konishi H, Komatsu S, Fujiwara H, Okamoto K, Otsuji E. The Role of cIAP1 and XIAP in Apoptosis Induced by Tumor Necrosis Factor Alpha in Esophageal Squamous Cell Carcinoma Cells. Dig Dis Sci 2017; 62:652-659. [PMID: 28050781 DOI: 10.1007/s10620-016-4430-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 12/20/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND The inhibitor of apoptosis protein (IAP) family are reported to play important roles in cancer cells evading apoptosis. However, the significance of their expression in human esophageal squamous cell carcinoma (ESCC) cells remains uncertain. AIMS The present study aimed to investigate the role of the IAP family members in tumor necrosis factor-α (TNF-α)-induced apoptosis of human ESCC cells. METHODS Five human ESCC cell lines were pretreated with TNF-α, cycloheximide (CHX, protein synthesis inhibitor), epoxomicin (proteasome inhibitor). Apoptosis assay and protein study with Western blot testing were conducted. Knockdown experiments with IAP siRNA were conducted, and the effect on cell apoptosis was analyzed. RESULTS Significant apoptosis was induced in five ESCC cell lines by TNF-α plus CHX stimulation, but not when treated with TNF-α or CHX alone. The protein expression levels of cIAP1 and XIAP were decreased by treatment with TNF-α in the presence of CHX, and the degree of cIAP1 and XIAP expression decrease was correlated with sensitivity to TNF-α plus CHX-induced apoptosis. Epoxomicin suppressed TNF-α plus CHX-induced degradation of survivin, cIAP1, and XIAP, in addition to apoptosis. A caspase inhibitor (z-VAD-fmk) suppressed TNF-α plus CHX-induced apoptosis, but did not suppress degradation of survivin, cIAP1, and XIAP. Furthermore, cIAP1 or XIAP siRNA transfected cells underwent apoptosis in response to treatment with TNF-α alone. Double knockdown of both genes resulted in further increased apoptosis. CONCLUSION cIAP1 and XIAP play an essential role in the resistance of ESCC cells against apoptosis.
Collapse
Affiliation(s)
- Shoichiro Hikami
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Maki Kitagawa-Juge
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
17
|
Impact of inhibitor of apoptosis proteins on immune modulation and inflammation. Immunol Cell Biol 2016; 95:236-243. [DOI: 10.1038/icb.2016.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022]
|
18
|
Abstract
Chronic kidney disease (CKD) is a lethal and rapidly increasing burden on society. Despite this, there are relatively few therapies in development for the treatment of CKD. Several recent costly phase 3 trials have failed to provide improved renal outcomes, diminishing interest in pharmaceutical investment. Furthermore, poor patient, physician, and payer awareness of CKD as a diagnosis has contributed to slow trial enrollment and successful implementation of these trials. Nevertheless, several therapeutics remain in development for the treatment of CKD, including mineralocorticoid-receptor antagonists, sodium/glucose cotransporter 2 inhibitors, anti-inflammatory drugs, and drugs that mitigate oxidative injury. Success of future CKD therapeutic trials will depend not only on improved understanding of disease pathogenesis, but also on improved trial enrollment rates, through increasing awareness of this disease by the public, policy makers, and the greater medical community.
Collapse
Affiliation(s)
- Matthew D Breyer
- Biotechnology Discovery Research, Eli Lilly and Company, Indianapolis, IN.
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
19
|
Pleiotropic properties of ASK1. Biochim Biophys Acta Gen Subj 2016; 1861:3030-3038. [PMID: 27693599 DOI: 10.1016/j.bbagen.2016.09.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/16/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Apoptosis signal-regulating kinase 1 (ASK1), also known as mitogen-activated protein kinase kinase kinase 5 (MAP3K5), has the potential to induce cellular apoptosis under various physiological conditions. It has long been suggested that ASK1 is highly sensitive to oxidative stress and contributes substantially to apoptosis. However, recent studies have indicated that ASK1 has pleiotropic roles in living organisms through other mechanisms in addition to apoptosis. SCOPE OF THE REVIEW This review describes the physiological functions of ASK1 in living organisms, focusing on the regulatory mechanisms of ASK1 activity and its importance in the pathogenesis of various diseases. We also highlight recent works published within the past few years. MAJOR CONCLUSIONS ASK1 forms a high-molecular-mass complex within the cell, designated as the ASK1 signalosome. Soon after the discovery of ASK1, several regulatory components of the ASK1 signalosome have been revealed, including thioredoxin (Trx), tumor-necrosis factor α receptor-associated factors (TRAFs) and 14-3-3s. In parallel with the precise analyses unveiling the molecular basis of ASK1 regulation, the physiological or pathophysiological significance of ASK1 in diverse organs has been elucidated. In addition to the generation of global knockout mice or tissue-specific knockout mice, ASK1-specific inhibitors have illuminated the biological roles of ASK1. GENERAL SIGNIFICANCE The multi-faceted features of the function of ASK1 have been discovered over the past two decades, revealing that ASK1 is a crucial molecule for maintaining cellular homeostasis, especially under conditions of stress. Based on the results that ASK1 deficiency provides beneficial effects for several diseases, modulating ASK1 activity is a promising method to ameliorate a subset of diseases.
Collapse
|
20
|
Abstract
Chronic kidney disease (CKD) represents a leading cause of death in the United States. There is no cure for this disease, with current treatment strategies relying on blood pressure control through blockade of the renin-angiotensin system. Such approaches only delay the development of end-stage kidney disease and can be associated with serious side effects. Recent identification of several novel mechanisms contributing to CKD development - including vascular changes, loss of podocytes and renal epithelial cells, matrix deposition, inflammation and metabolic dysregulation - has revealed new potential therapeutic approaches for CKD. This Review assesses emerging strategies and agents for CKD treatment, highlighting the associated challenges in their clinical development.
Collapse
|
21
|
Yu Z, Chen T, Li X, Yang M, Tang S, Zhu X, Gu Y, Su X, Xia M, Li W, Zhang X, Wang Q, Cao X, Wang J. Lys29-linkage of ASK1 by Skp1-Cullin 1-Fbxo21 ubiquitin ligase complex is required for antiviral innate response. eLife 2016; 5. [PMID: 27063938 PMCID: PMC4887211 DOI: 10.7554/elife.14087] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/09/2016] [Indexed: 12/25/2022] Open
Abstract
Protein ubiquitination regulated by ubiquitin ligases plays important roles in innate immunity. However, key regulators of ubiquitination during innate response and roles of new types of ubiquitination (apart from Lys48- and Lys63-linkage) in control of innate signaling have not been clearly understood. Here we report that F-box only protein Fbxo21, a functionally unknown component of SCF (Skp1–Cul1–F-box protein) complex, facilitates Lys29-linkage and activation of ASK1 (apoptosis signal-regulating kinase 1), and promotes type I interferon production upon viral infection. Fbxo21 deficiency in mice cells impairs virus-induced Lys29-linkage and activation of ASK1, attenuates c-Jun N-terminal kinase (JNK) and p38 signaling pathway, and decreases the production of proinflammatory cytokines and type I interferon, resulting in reduced antiviral innate response and enhanced virus replication. Therefore Fbxo21 is required for ASK1 activation via Lys29-linkage of ASK1 during antiviral innate response, providing mechanistic insights into non-proteolytic roles of SCF complex in innate immune response. DOI:http://dx.doi.org/10.7554/eLife.14087.001 The innate immune system is the body’s first line of defense against being infected by viruses and other microbes. Upon recognizing a virus, host cells trigger the innate immune response in an effort to eliminate the threat. However, innate immune responses must be carefully controlled because an excessive response can cause inflammation that harms the body. The innate immune response involves a variety of cells and processes that are each activated through a series of communication systems called signaling pathways. While much has been learned about which parts of a virus trigger the innate immune response, it is not clear how the immune response to the virus is controlled. It has been suggested that a process known as ubiquitination could be involved in regulating the activity of signaling pathways that activate the innate immune response. During ubiquitination, enzymes attach a small molecule called ubiquitin to a specific target protein. Ubiquitin often acts as a label that targets a particular protein for destruction. Enzymes called E3 ubiquitin ligases play central roles in identifying specific target proteins for ubiquitination. Some of these enzymes consist of a single protein unit that acts alone, but other E3 ubiquitin ligases are formed by groups (or “complexes”) of several proteins working together. Members of the F-box only protein family are components of some ubiquitin ligase complexes. Here, Yu et al. used a “microarray” technique to assess which F-box only proteins in mice are produced during an immune response to two viruses. The experiments identified an F-box protein called Fbxo21 as a potential candidate for a role in regulating the innate immune response. Additional experiments revealed that Fbxo21 is involved in adding ubiquitin to a specific location on a signaling protein called ASK1, which is known to be crucial for innate immune responses. Instead of targeting ASK1 for destruction, this ubiquitination activates ASK1. Therefore, Yu et al.’s findings demonstrate that Fbxo21 plays an important role in regulating innate immune responses. A future challenge is to investigate exactly how ASK1 is activated by the ubiquitin. DOI:http://dx.doi.org/10.7554/eLife.14087.002
Collapse
Affiliation(s)
- Zhou Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China.,National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Taoyong Chen
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xuelian Li
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Mingjin Yang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China.,National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Songqing Tang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuhui Zhu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yan Gu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xiaoping Su
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Meng Xia
- National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihua Li
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Xuemin Zhang
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, China.,National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianli Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Zhang QS, Eaton GJ, Diallo C, Freeman TA. Stress-Induced Activation of Apoptosis Signal-Regulating Kinase 1 Promotes Osteoarthritis. J Cell Physiol 2015; 231:944-53. [PMID: 26405834 DOI: 10.1002/jcp.25186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/03/2015] [Indexed: 12/11/2022]
Abstract
Apoptosis signal-regulated kinase 1 (ASK1) has been shown to affect a wide range of cellular processes including stress-related responses, cytokine and growth factor signaling, cell cycle and cell death. Recently, we reported that lack of ASK1 slowed chondrocyte hypertrophy, terminal differentiation and apoptosis resulting in an increase in trabecular bone formation. Herein, we investigated the role of ASK1 in the pathogenesis of osteoarthritis (OA). Immunohistochemistry performed on articular cartilage samples from patients with OA showed ASK1 expression increased with OA severity. In vitro analysis of chondrocyte hypertrophy, maturation and ASK1 signaling in embryonic fibroblasts from ASK1 knockout (KO) and wild type (WT) mice was examined. Western analysis demonstrated an increase in ASK1 signaling commensurate with chondrogenic maturation during differentiation or in response to stress by the cytokines, tumor necrosis factor alpha or interleukin 1 beta in WT, but not in ASK1 KO embryonic fibroblasts. Surgically induced moderate or severe OA or OA due to natural aging in WT and ASK1 KO mice was assessed by microCT of subchondral bone, immunohistochemistry, histology, and OARSI scoring. Immunohistochemistry, microCT and OARSI scoring all indicated that the lack of ASK1 protected against OA joint degeneration, both in surgically induced OA and in aging mice. We propose that the ASK1 MAP kinase signaling cascade is an important regulator of chondrocyte terminal differentiation and inhibitors of this pathway could be useful for slowing chondrocyte maturation and cell death observed with OA progression. J. Cell. Physiol. 231: 944-953, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qian-Shi Zhang
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Spine Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Gregory J Eaton
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Carol Diallo
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Theresa A Freeman
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
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
|
24
|
Jiang WL, Zhang YF, Xia QQ, Zhu J, Yu X, Fan T, Wang F. MicroRNA-19a regulates lipopolysaccharide-induced endothelial cell apoptosis through modulation of apoptosis signal-regulating kinase 1 expression. BMC Mol Biol 2015; 16:11. [PMID: 25982447 PMCID: PMC4446110 DOI: 10.1186/s12867-015-0034-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/19/2015] [Indexed: 01/07/2023] Open
Abstract
Background MicroRNAs, small non-encoding RNAs that post-transcriptionally modulate expression of their target genes, have been implicated as critical regulatory molecules in endothelial cells. Results In the present study, we found that overexpression of miR-19a protects endothelial cells from lipopolysaccharide (LPS)-induced apoptosis through the apoptosis signal-regulating kinase 1 (ASK1)/p38 pathway. Quantitative real-time PCR demonstrated that the expression of miR-19a in endothelial cell was markedly down-regulated by LPS stimulation. Furthermore, LPS-induced apoptosis was significantly inhibited by over-expression of miR-19a. Finally, both a luciferase reporter assay and western blot analysis showed that ASK1 is a direct target of miR-19a. Conclusions MiR-19a regulates ASK1 expression by targeting specific binding sites in the 3’ untranslated region of ASK1 mRNA. Overexpression of miR-19a is an effective method to protect against LPS-induced apoptosis of endothelial cells. Electronic supplementary material The online version of this article (doi:10.1186/s12867-015-0034-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Long Jiang
- Department of Respiration, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Yu-Feng Zhang
- Department of Respiration, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Qing-Qing Xia
- Department of Respiration, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Jian Zhu
- Department of Neurology, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Xin Yu
- Department of Internal Medicine, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Tao Fan
- Department of Neurology, Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin City, Jiangsu Province, 214400, China.
| | - Feng Wang
- Department of Neurology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
25
|
Tsai YC, Teng YN, Hung JH, Wu CH, Kuo YT, Kuo PL, Chiu CC, Huang B. Correlation between leucine rich domain and the stability of LRWD1 protein in human NT2/D1 cells. Adv Med Sci 2014; 59:266-72. [PMID: 25170821 DOI: 10.1016/j.advms.2014.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/13/2014] [Accepted: 07/23/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE LRWD1 is a protein that contains LRR and WDs domains and is important in regulating spermatogenesis. However, the roles of LRR or WDs domains in the expression of LRWD1 remain unclear. MATERIALS AND METHODS The NT2/D1 cells separately transfected with full length of LRWD1 gene (LRWD(WT)) or genes with deleted sequences in the LRR domain (LRWD1(ΔLRR)), WD1 domain (LRWD1(ΔWD1)), WD2 domain (LRWD1(ΔWD2)), WD3 domain (LRWD1(ΔWD3)) and entire three WD domains (LRWD1(Δ3×WD)) were applied to investigate the expression levels of LRWD1 protein by either Western blot or flow cytometry. The associated proteins in these mutated LRWD1 proteins were identified by mass spectrometry. RESULTS Deletion of the LRR domain significantly decreased the expression of LRWD1 protein. With the treatment of MG132, the LRR domain may functions in preventing LRWD1 protein from proteasome-mediated degradation. In the co-immunoprecipitation analysis, protein receptor of tumor necrosis factor 2 (TNFR2) was specifically observed to be associated with LRR-deficient LRWD1 protein. CONCLUSIONS The LRR domain is significantly correlated to the stability of LRWD1 protein. Determining if the stability is modulated by TNFR2 is worthy of further study.
Collapse
|
26
|
Effect of chemical modification on the ability of pyrrolidinium fullerene to induce apoptosis of cells transformed by JAK2 V617F mutant. Int Immunopharmacol 2014; 20:258-63. [PMID: 24631513 DOI: 10.1016/j.intimp.2014.02.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/26/2014] [Accepted: 02/26/2014] [Indexed: 11/22/2022]
Abstract
JAK2 V617F mutant, a gene responsible for human myeloproliferative neoplasms (MPNs), causes not only cellular transformation but also resistance to various anti-cancer drugs. We previously reported that pyrrolidinium fullerene markedly induced the apoptosis of JAK2 V617F mutant-induced transformed cells through the reduction of apoptosis signal-regulating kinase 1 (ASK1), following inhibition of the c-Jun N-terminal kinase (JNK) pathway. In the current study, we found that the replacement of the 2-hydrogen atom (H) or N-methyl group (CH3) by the butyl group (C4C9) caused the more than 3-fold potent cytotoxic effects on cells transformed by the JAK2 V617F mutant. Strikingly, these chemical modification of pyrrolidinium fullerene resulted in more marked reduction of ASK1 protein and a more potent inhibitory effect on the JNK signaling cascade. On the other hand, when modified with a longer alkyl group, the derivatives lacked their cytotoxicity. These observations clearly indicate that the modification of pyrrolidinium fullerene with a suitable length of alkyl group such as butyl group enhances its apoptotic effect through inhibition of the ASK1-MKK4/7-JNK pathway.
Collapse
|
27
|
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
|
28
|
Ruspi G, Schmidt EM, McCann F, Feldmann M, Williams RO, Stoop AA, Dean JLE. TNFR2 increases the sensitivity of ligand-induced activation of the p38 MAPK and NF-κB pathways and signals TRAF2 protein degradation in macrophages. Cell Signal 2013; 26:683-90. [PMID: 24378531 DOI: 10.1016/j.cellsig.2013.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/19/2013] [Accepted: 12/22/2013] [Indexed: 12/23/2022]
Abstract
Tumour necrosis factor (p55 or p60) receptor (TNFR) 1 is the major receptor that activates pro-inflammatory signalling and induces gene expression in response to TNF. Consensus is lacking for the function of (p75 or p80) TNFR2 but experiments in mice have suggested neuro-, cardio- and osteo-protective and anti-inflammatory roles. It has been shown in various cell types to be specifically required for the induction of TNFR-associated factor-2 (TRAF2) degradation and activation of the alternative nuclear factor (NF)-kappaB pathway, and to contribute to the activation of mitogen-activated protein kinases (MAPK) and the classical NF-kappaB pathway. We have investigated the signalling functions of TNFR2 in primary human and murine macrophages. We find that in these cells TNF induces TRAF2 degradation, and this is blocked in TNFR2(-/-) macrophages. TRAF2 has been previously reported to be required for TNF-induced activation of p38 MAPK. However, TRAF2 degradation does not inhibit TNF-induced tolerance of p38 MAPK activation. Neither TNF, nor lipopolysaccharide treatment, induced activation of the alternative NF-kappaB pathway in macrophages. Activation by TNF of the p38 MAPK and NF-kappaB pathways was blocked in TNFR1(-/-) macrophages. In contrast, although TNFR2(-/-) macrophages displayed robust p38 MAPK activation and IkappaBα degradation at high concentrations of TNF, at lower doses the concentration dependence of signalling was weakened by an order of magnitude. Our results suggest that, in addition to inducing TRAF2 protein degradation, TNFR2 also plays a crucial auxiliary role to TNFR1 in sensitising macrophages for the ligand-induced activation of the p38 MAPK and classical NF-kappaB pro-inflammatory signalling pathways.
Collapse
Affiliation(s)
- Gerhard Ruspi
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Emily M Schmidt
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Fiona McCann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Richard O Williams
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - A Allart Stoop
- Innovation Biopharm Discovery Unit, Biopharm R&D, GlaxoSmithKline, Cambridge CB4 0WG, United Kingdom
| | - Jonathan L E Dean
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
29
|
Tumor necrosis factor receptor 2: its contribution to acute cellular rejection and clear cell renal carcinoma. BIOMED RESEARCH INTERNATIONAL 2013; 2013:821310. [PMID: 24350291 PMCID: PMC3848079 DOI: 10.1155/2013/821310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 12/26/2022]
Abstract
Tumor necrosis factor receptor 2 (TNFR2) is a type I transmembrane glycoprotein and one of the two receptors that orchestrate the complex biological functions of tumor necrosis factor (TNF, also designed TNF-α). Accumulating experimental evidence suggests that TNFR2 plays an important role in renal disorders associated with acute cellular rejection and clear cell renal carcinoma but its exact role in these settings is still not completely understood. This papers reviews the factors that may mediate TNFR2 induction in acute cellular rejection and clear cell renal carcinoma and its contribution to these conditions and discusses its therapeutic implications. A greater understanding of the function of TNFR2 may lead to the development of new anti-TNF drugs.
Collapse
|
30
|
Waters JP, Pober JS, Bradley JR. Tumour necrosis factor in infectious disease. J Pathol 2013; 230:132-47. [PMID: 23460469 DOI: 10.1002/path.4187] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 02/15/2013] [Accepted: 02/23/2013] [Indexed: 12/12/2022]
Abstract
TNF signals through two distinct receptors, designated TNFR1 and TNFR2, which initiate diverse cellular effects that include cell survival, activation, differentiation, and proliferation and cell death. These cellular responses can promote immunological and inflammatory responses that eradicate infectious agents, but can also lead to local tissue injury at sites of infection and harmful systemic effects. Defining the molecular mechanisms involved in TNF responses, the effects of natural and experimental genetic diversity in TNF signalling and the effects of therapeutic blockade of TNF has increased our understanding of the key role that TNF plays in infectious disease.
Collapse
Affiliation(s)
- John P Waters
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | | | |
Collapse
|
31
|
Giardino Torchia ML, Conze DB, Ashwell JD. c-IAP1 and c-IAP2 redundancy differs between T and B cells. PLoS One 2013; 8:e66161. [PMID: 23799077 PMCID: PMC3684576 DOI: 10.1371/journal.pone.0066161] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/02/2013] [Indexed: 01/02/2023] Open
Abstract
Cellular Inhibitors of Apoptosis 1 and 2 (c-IAP1 and c-IAP2) are ubiquitin protein ligases (E3s) that constitutively ubiquitinate and induce proteasomal-mediated degradation of NF-κB Inducing Kinase (NIK) and repress non-canonical NF-κB activation. Mice expressing an E3-inactive c-IAP2 mutant (c-IAP2(H570A)) have constitutive activation of non-canonical NF-κB, resulting in B cell hyperplasia and T cell costimulation-independence. If, and if so to what extent, c-IAP1 and c-IAP2 are redundant in NF-κB regulation in these mice is not known. Here we have generated mice expressing a mutant c-IAP1 that lacks E3 activity (c-IAP1(H582A)). These mice were phenotypically normal and did not have constitutive NF-κB activation in B cells or MEFs. siRNA-mediated knockdown of c-IAP2 showed that accumulated c-IAP2, resulting from lack of c-IAP1-dependent degradation, compensated for absent c-IAP1 E3 activity. Surprisingly, c-IAP1(H582A) T cells had a lower p100/p52 ratio than wild type T cells, and in the absence of costimulation proliferated to a degree intermediate between wild type and c-IAP2(H570A) T cells. Therefore, although c-IAP1 and c-IAP2 both can repress constitutive NF-κB activation, the relative importance of each varies according to cell type.
Collapse
Affiliation(s)
- Maria Letizia Giardino Torchia
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dietrich B. Conze
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
Philippe L, Alsaleh G, Pichot A, Ostermann E, Zuber G, Frisch B, Sibilia J, Pfeffer S, Bahram S, Wachsmann D, Georgel P. MiR-20a regulates ASK1 expression and TLR4-dependent cytokine release in rheumatoid fibroblast-like synoviocytes. Ann Rheum Dis 2013; 72:1071-9. [PMID: 23087182 DOI: 10.1136/annrheumdis-2012-201654] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To evaluate whether miR-20a belonging to the cluster miR-17-92 is a negative regulator of inflammation in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) by modulating expression of apoptosis signal-regulating kinase (ASK) 1, a key component of the toll-like receptors 4 pathway, upstream of p38 mitogen-activated protein kinase. METHODS Evaluation of miR-20a and ASK1 mRNA was performed by RT-qPCR. ASK1 protein expression was assessed by western blotting. Overexpression of miR-20a was performed by transfection of RA FLS and THP-1 cells with miR-20a mimics. Interleukin (IL)-6, CXCL-10, IL-1β and TNF-α release were measured by ELISA. The role of miR-20a in vivo was assessed by IL-6 release from macrophages obtained from mice injected intraperitoneally with vectorised miR-20a mimics. RESULTS We showed that stimulation of RA FLS with lipopolysacharide (LPS) and bacterial lipoproteins (BLP) induces a drop in expression of miR-20a and that this decrease is associated with an upregulation of ASK1 expression. Using transfection of Ask1 3'UTR reporters, we demonstrate that Ask1 is a direct target of miR-20a. Overexpression of miR-20a led to a global decrease in ASK1 protein in BLP- and LPS-activated cells indicating that miR-20a regulates the expression of ASK1 at the translational level. Transfection of miR-20a mimics decreases IL-6 and CXCL10 release by RA FLS and IL-1β and TNF-α by activated THP-1 cells but only in response to LPS. Last, injection of vectorised miR-20a mimics to mice led to a global decrease in ASK1 protein expression and IL-6 secretion in LPS-activated macrophages. CONCLUSIONS Our data point toward an important role for miR-20a in the regulation of pro-inflammatory cytokines release, by controlling ASK1 expression in RA FLS.
Collapse
Affiliation(s)
- Lucas Philippe
- ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Université de Strasbourg, Strasbourg, Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Philippe L, Alsaleh G, Bahram S, Pfeffer S, Georgel P. The miR-17 ∼ 92 Cluster: A Key Player in the Control of Inflammation during Rheumatoid Arthritis. Front Immunol 2013; 4:70. [PMID: 23516027 PMCID: PMC3601326 DOI: 10.3389/fimmu.2013.00070] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/04/2013] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are now recognized as essential regulators of gene expression in plants and animals. They potentially modulate the expression of multiple genes thereby enabling homeostatic settings in physiological conditions. Their role is also increasingly considered in many diseases in which deregulated epigenetic mechanisms induce aberrant gene expression. Work conducted in our laboratory has recently led to the identification of miRNAs essential for the control of inflammatory reactions that occur during rheumatoid arthritis (RA). In this review, we describe two such miRNAs, members of the miR-17 ∼ 92 cluster, which has been previously implicated in cancer. Based on our data and on predicted miRNA:mRNA interactions, we will extrapolate a model whereby the miR-17 ∼ 92 cluster appears as a global regulator of the Apoptosis Signal-Regulating Kinase 1 signalosome, a central actor in the inflammatory pathways activated during RA. We will also discuss the potential therapeutic outcomes emerging from this model.
Collapse
Affiliation(s)
- Lucas Philippe
- ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Centre de Recherche d'Immunologie et d'Hématologie, Université de Strasbourg Strasbourg Cedex, France
| | | | | | | | | |
Collapse
|
34
|
Tan BM, Zammit NW, Yam AO, Slattery R, Walters SN, Malle E, Grey ST. Baculoviral inhibitors of apoptosis repeat containing (BIRC) proteins fine-tune TNF-induced nuclear factor κB and c-Jun N-terminal kinase signalling in mouse pancreatic beta cells. Diabetologia 2013; 56:520-32. [PMID: 23250032 DOI: 10.1007/s00125-012-2784-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 10/19/2012] [Indexed: 10/27/2022]
Abstract
AIMS/HYPOTHESIS For beta cells, contact with TNF-α triggers signalling cascades that converge on pathways important for cell survival and inflammation, specifically nuclear factor κB (NF-κB), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase pathways. Here, we investigated the function of baculoviral inhibitors of apoptosis repeat containing (BIRC) proteins in regulating TNF signalling cascades. METHODS TNF regulation of Birc genes was studied by mRNA expression and promoter analysis. Birc gene control of cell signalling was studied in beta cell lines, and in islets from Birc2(-/-) and Birc3(-/-) mice, and from Birc3(-/-) Birc2Δ beta cell mice that selectively lack Birc2 and Birc3 (double knockout [DKO]). Islet function was tested by intraperitoneal glucose tolerance test and transplantation. RESULTS TNF-α selectively induced Birc3 in beta cells, which in turn was sufficient to drive and potentiate NF-κB reporter activity. Conversely, Birc3(-/-) islets exhibited delayed TNF-α-induced IκBα degradation with reduced expression of Ccl2 and Cxcl10. DKO islets showed a further delay in IκBα degradation kinetics. Surprisingly, DKO islets exhibited stimulus-independent and TNF-dependent hyperexpression of TNF target genes A20 (also known as Tnfaip3), Icam1, Ccl2 and Cxcl10. DKO islets showed hyperphosphorylation of the JNK-substrate, c-Jun, while a JNK-antagonist prevented increases of Icam1, Ccl2 and Cxcl10 expression. Proteosome blockade of MIN6 cells phenocopied DKO islets. DKO islets showed more rapid loss of glucose homeostasis when challenged with the inflammatory insult of transplantation. CONCLUSIONS/INTERPRETATION BIRC3 provides a feed-forward loop, which, with BIRC2, is required to moderate the normal speed of NF-κB activation. Paradoxically, BIRC2 and BIRC3 act as a molecular brake to rein in activation of the JNK signalling pathway. Thus BIRC2 and BIRC3 fine-tune NF-κB and JNK signalling to ensure transcriptional responses are appropriately matched to extracellular inputs. This control is critical for the beta cell's stress response.
Collapse
Affiliation(s)
- B M Tan
- Gene Therapy and Autoimmunity Group, Immunology Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
| | | | | | | | | | | | | |
Collapse
|
35
|
Yang W, Yang X, David G, Dorsey JF. Dissecting the complex regulation of Mad4 in glioblastoma multiforme cells. Cancer Biol Ther 2012; 13:1339-48. [PMID: 22895069 DOI: 10.4161/cbt.21814] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Among proteins in the c-Myc/Max/Mad/Sin3 regulatory complex, Mad4 and Sin3B are routinely detected in human glioblastoma multiforme (GBM) cell lines. In response to gamma radiation, the expression of Sin3B and Mad4 in GBM cells was upregulated in parallel over time, suggesting that Sin3B may play a role in the regulation of Mad4 stability. In agreement with this hypothesis, exogenously expressed Sin3B significantly stabilized co-transfected Mad4 and, to a lesser extent, endogenous Mad4. In addition, siRNA silencing of Sin3B induced an increase in the expression of c-Myc and Sin3A, which contributed to increased expression of Mad4. Simultaneous silencing of Sin3B, Sin3A and c-Myc decreased Mad4 stability to a greater extent than silencing of Sin3B alone. Although Mad1 was reported to be a target of c-IAP1 E3 ligase activity for degradation, the E3 ligase activity of c-IAP1 was not required for downregulation of Mad4 expression. The association of c-IAP1 with Sin3B or Mad4 suggested that Sin3B might interfere with the binding of c-IAP1 to Mad4; however, overexpression of Sin3B did not affect the interaction between Mad4 and c-IAP1. Instead, direct binding of Sin3B to c-IAP1 may protect Mad4 from degradation by c-IAP1, leading to enhanced stability of Mad4. Exogenous expression of Sin3B also inhibited c-IAP1-mediated degradation of Mad1, TRAF2, c-IAP2 and ASK1, known targets of c-IAP1 E3 ligase activity. These results indicate that Sin3B, together with other c-Myc regulatory members, maintain the steady-state level of Mad4, in part through inhibition of c-IAP1-mediated degradation of Mad4.
Collapse
Affiliation(s)
- Wensheng Yang
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
36
|
Funakoshi-Tago M, Nagata T, Tago K, Tsukada M, Tanaka K, Nakamura S, Mashino T, Kasahara T. Fullerene derivative prevents cellular transformation induced by JAK2 V617F mutant through inhibiting c-Jun N-terminal kinase pathway. Cell Signal 2012; 24:2024-34. [PMID: 22750290 DOI: 10.1016/j.cellsig.2012.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/25/2012] [Indexed: 10/28/2022]
Abstract
The constitutively activated mutation (V617F) of tyrosine kinase Janus kinase 2 (JAK2) is found in the majority of patients with myeloproliferative neoplasms (MPNs). The development of a novel chemical compound to suppress JAK2 V617F mutant-induced onset of MPNs and clarification of the signaling cascade downstream of JAK2 V617F mutant will provide clues to treat MPNs. Here we found that a water-soluble pyrrolidinium fullerene derivative, C(60)-bis (N, N-dimethylpyrrolidinium iodide), markedly induced apoptosis of JAK2 V617F mutant-induced transformed cells through a novel mechanism, inhibiting c-Jun N-terminal kinase (JNK) activation pathway but not generation of reactive oxygen species (ROS). Pyrrolidinium fullerene derivative significantly reduced the protein expression level of apoptosis signal-regulating kinase 1 (ASK1), one of the mitogen-activated protein kinase kinase kinases (MAPKKK), resulting in the inhibition of upstream molecules of JNK, mitogen-activated protein kinase kinase 4 (MKK4) and mitogen-activated protein kinase kinase 7 (MKK7). Strikingly, the knockdown of ASK1 enhanced the sensitivity to pyrrolidinium fullerene derivative-induced apoptosis, and the treatment with a JNK inhibitor, SP600125, also induced apoptosis of the transformed cells by JAK2 V617F mutant. Furthermore, administration of both SP600125 and pyrrolidinium fullerene derivative markedly inhibited JAK2 V617F mutant-induced tumorigenesis in nude mice. Taking these findings together, JAK2 V617F mutant-induced JNK signaling pathway is an attractive target for MPN therapy, and pyrrolidinium fullerene derivative is now considered a candidate potent drug for MPNs.
Collapse
|
37
|
Bluml S, Scheinecker C, Smolen JS, Redlich K. Targeting TNF receptors in rheumatoid arthritis. Int Immunol 2012; 24:275-81. [DOI: 10.1093/intimm/dxs047] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
38
|
Flygare JA, Beresini M, Budha N, Chan H, Chan IT, Cheeti S, Cohen F, Deshayes K, Doerner K, Eckhardt SG, Elliott LO, Feng B, Franklin MC, Reisner SF, Gazzard L, Halladay J, Hymowitz SG, La H, LoRusso P, Maurer B, Murray L, Plise E, Quan C, Stephan JP, Young SG, Tom J, Tsui V, Um J, Varfolomeev E, Vucic D, Wagner AJ, Wallweber HJA, Wang L, Ware J, Wen Z, Wong H, Wong JM, Wong M, Wong S, Yu R, Zobel K, Fairbrother WJ. Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J Med Chem 2012; 55:4101-13. [PMID: 22413863 DOI: 10.1021/jm300060k] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A series of compounds were designed and synthesized as antagonists of cIAP1/2, ML-IAP, and XIAP based on the N-terminus, AVPI, of mature Smac. Compound 1 (GDC-0152) has the best profile of these compounds; it binds to the XIAP BIR3 domain, the BIR domain of ML-IAP, and the BIR3 domains of cIAP1 and cIAP2 with K(i) values of 28, 14, 17, and 43 nM, respectively. These compounds promote degradation of cIAP1, induce activation of caspase-3/7, and lead to decreased viability of breast cancer cells without affecting normal mammary epithelial cells. Compound 1 inhibits tumor growth when dosed orally in the MDA-MB-231 breast cancer xenograft model. Compound 1 was advanced to human clinical trials, and it exhibited linear pharmacokinetics over the dose range (0.049 to 1.48 mg/kg) tested. Mean plasma clearance in humans was 9 ± 3 mL/min/kg, and the volume of distribution was 0.6 ± 0.2 L/kg.
Collapse
Affiliation(s)
- John A Flygare
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cartier J, Berthelet J, Marivin A, Gemble S, Edmond V, Plenchette S, Lagrange B, Hammann A, Dupoux A, Delva L, Eymin B, Solary E, Dubrez L. Cellular inhibitor of apoptosis protein-1 (cIAP1) can regulate E2F1 transcription factor-mediated control of cyclin transcription. J Biol Chem 2011; 286:26406-17. [PMID: 21653699 PMCID: PMC3143604 DOI: 10.1074/jbc.m110.191239] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 06/06/2011] [Indexed: 11/06/2022] Open
Abstract
The inhibitor of apoptosis protein cIAP1 (cellular inhibitor of apoptosis protein-1) is a potent regulator of the tumor necrosis factor (TNF) receptor family and NF-κB signaling pathways in the cytoplasm. However, in some primary cells and tumor cell lines, cIAP1 is expressed in the nucleus, and its nuclear function remains poorly understood. Here, we show that the N-terminal part of cIAP1 directly interacts with the DNA binding domain of the E2F1 transcription factor. cIAP1 dramatically increases the transcriptional activity of E2F1 on synthetic and CCNE promoters. This function is not conserved for cIAP2 and XIAP, which are cytoplasmic proteins. Chromatin immunoprecipitation experiments demonstrate that cIAP1 is recruited on E2F binding sites of the CCNE and CCNA promoters in a cell cycle- and differentiation-dependent manner. cIAP1 silencing inhibits E2F1 DNA binding and E2F1-mediated transcriptional activation of the CCNE gene. In cells that express a nuclear cIAP1 such as HeLa, THP1 cells and primary human mammary epithelial cells, down-regulation of cIAP1 inhibits cyclin E and A expression and cell proliferation. We conclude that one of the functions of cIAP1 when localized in the nucleus is to regulate E2F1 transcriptional activity.
Collapse
Affiliation(s)
- Jessy Cartier
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Jean Berthelet
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Arthur Marivin
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Simon Gemble
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Valérie Edmond
- Inserm U823, Equipe Bases Moléculaires de la Progression des Cancers du Poumon, Institut Albert Bonniot, Grenoble F-38042, France
- the Université Joseph Fourier, Grenoble, F-38041, France, and
| | - Stéphanie Plenchette
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Brice Lagrange
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Arlette Hammann
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Alban Dupoux
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Laurent Delva
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| | - Béatrice Eymin
- Inserm U823, Equipe Bases Moléculaires de la Progression des Cancers du Poumon, Institut Albert Bonniot, Grenoble F-38042, France
- the Université Joseph Fourier, Grenoble, F-38041, France, and
| | - Eric Solary
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
- Inserm UMR1009, Institut Gustave Roussy, Villejuif, F-94805, France
| | - Laurence Dubrez
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) UMR866, Dijon, F-21079, France
- the Faculty of Medicine, University of Burgundy, Institut Fédératif de Recherche (IFR) 100, Dijon, F-21079, France
| |
Collapse
|
40
|
Targeting inhibitor of apoptosis proteins for therapeutic intervention. Future Med Chem 2011; 1:1509-25. [PMID: 21426063 DOI: 10.4155/fmc.09.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inhibitors of apoptosis (IAP) proteins have emerged over the last decade as important targets for therapeutic intervention in human malignancies. Overexpression of IAPs has been implicated in cell survival and resistance against stress-induced apoptosis brought on by radiation and/or chemotherapeutics (currently the standard-of-care in a variety of different cancer diseases). In addition, evasion from death receptor-mediated apoptosis and regulation of NF-κB pathways and cell division have also been associated with IAP proteins. Efforts to target IAP proteins in tumors have focused mainly on designing small molecules that mimic the IAP-binding motif of the endogenous IAP antagonist, second mitochondrial activator of caspases. In addition, several other IAP-targeting strategies, including antisense oligonucleotides and transcriptional repression, have also been initiated, with the hope of providing therapeutic benefit to cancer patients.
Collapse
|
41
|
ZNAMENSKAYA LF, YEGOROVA YUYU, ZITNER SV. Mechanism of the biological effect of the tumor necrosis factor-аlpha at psoriasis. VESTNIK DERMATOLOGII I VENEROLOGII 2011. [DOI: 10.25208/vdv974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Among different cytokines, tumor necrosis factor-аlpha (tumor necrosis factor α, TNF-α) plays a special role in psoriatic immunopathogenesis. Data on this cytokine collected for the recent decades made it possible to create a number of biological drugs blocking TNF-α, which are successfully applied in clinical practice for treating medium to severe psoriasis and psoriatic arthritis. This review presents general information about the cytokine structure and its receptor apparatus, regulation mechanisms of TNF-α synthesis and ways of signal transmission as the basis needed to implement the biological effects of cytokine in the development of psoriatic skin affections.
Collapse
|
42
|
Abstract
Extensive research has been performed to unravel the mechanistic signaling pathways mediated by tumor necrosis factor receptor 1 (TNFR1), by contrast there is limited knowledge on cellular signaling upon activation of TNFR2. Recently published data have revealed that these two receptors not only function independently, but also can influence each other via cross-talk between the different signaling pathways initiated by TNFR1 and TNFR2 stimulation. Furthermore, the complexity of this cross-talk is also dependent on the different signaling kinetics between TNFR1 and TNFR2, by which a delicate balance between cell survival and apoptosis can be maintained. Some known signaling factors and the kinetics that are involved in the receptor cross-talk between TNFR1 and TNFR2 are the topic of this review.
Collapse
Affiliation(s)
- Petrus J W Naudé
- Department of Molecular Neurobiology, University of Groningen, The Netherlands
| | | | | | | |
Collapse
|
43
|
Conze DB, Zhao Y, Ashwell JD. Non-canonical NF-κB activation and abnormal B cell accumulation in mice expressing ubiquitin protein ligase-inactive c-IAP2. PLoS Biol 2010; 8:e1000518. [PMID: 21048983 PMCID: PMC2964333 DOI: 10.1371/journal.pbio.1000518] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 09/02/2010] [Indexed: 11/19/2022] Open
Abstract
Chromosomal translocations between loci encoding MALT1 and c-IAP2 are common in MALT lymphomas. The resulting fusion proteins lack the c-IAP2 RING domain, the region responsible for its ubiquitin protein ligase (E3) activity. Ectopic expression of the fusion protein activates the canonical NF-κB signaling cascade, but how it does so is controversial and how it promotes MALT lymphoma is unknown. Considering recent reports implicating c-IAP1 and c-IAP2 E3 activity in repression of non-canonical NF-κB signaling, we asked if the c-IAP2/MALT fusion protein can initiate non-canonical NF-κB activation. Here we show that in addition to canonical activation, the fusion protein stabilizes NIK and activates non-canonical NF-κB. Canonical but not non-canonical activation depended on MALT1 paracaspase activity, and expression of E3-inactive c-IAP2 activated non-canonical NF-κB. Mice in which endogenous c-IAP2 was replaced with an E3-inactive mutant accumulated abnormal B cells with elevated non-canonical NF-κB and had increased numbers of B cells with a marginal zone phenotype, gut-associated lymphoid hyperplasia, and other features of MALT lymphoma. Thus, the c-IAP2/MALT1 fusion protein activates NF-κB by two distinct mechanisms, and loss of c-IAP2 E3 activity in vivo is sufficient to induce abnormalities common to MALT lymphoma.
Collapse
Affiliation(s)
- Dietrich B. Conze
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yongge Zhao
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
44
|
Abstract
Members of the nuclear factor-κB (NF-κB) family of transcription factors play critical roles in regulating the expression of genes whose products are involved in inflammation, the immune response, cell proliferation, and the suppression of both death receptor- and stress-induced apoptosis. Abnormal NF-κB activation has been observed in various inflammatory diseases and many types of cancers. Gene knockout studies have clearly demonstrated that most of the physiologically relevant stimuli that activate NF-κB converge on inhibitor of κB kinase (IKK). Although the mechanism by which IKK activates NF-κB is well established, the upstream signaling mechanisms-those that underlie IKK activation by IKK kinases (IKK-Ks)-are not yet fully understood. The current belief is that members of the TNF receptor-associated factor (TRAF) family function as ubiquitin E3 ligases that catalyze non-canonical polyubiquitination of adaptor proteins, and that the ubiquitinated adaptor proteins in turn serve as platforms to recruit IKK and IKK-Ks, facilitating IKK activation through proximity-mediated phosphorylation. This review will focus on the most recent findings relating to the role of TRAFs-mediated protein ubiquitination in regulating IKK activation, and highlight the newly emerging complexity of protein ubiquitination in receptor-induced NF-κB activation.
Collapse
Affiliation(s)
- Hasem Habelhah
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
45
|
|
46
|
Won M, Park KA, Byun HS, Sohn KC, Kim YR, Jeon J, Hong JH, Park J, Seok JH, Kim JM, Yoon WH, Jang IS, Shen HM, Liu ZG, Hur GM. Novel anti-apoptotic mechanism of A20 through targeting ASK1 to suppress TNF-induced JNK activation. Cell Death Differ 2010; 17:1830-41. [PMID: 20448643 DOI: 10.1038/cdd.2010.47] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The zinc-finger protein A20 has crucial physiological functions as a dual inhibitor of nuclear factor-κB (NF-κB) activation and apoptosis in tumor necrosis factor (TNF) receptor 1 signaling pathway. Although the molecular basis for the anti-NF-κB function of A20 has been well elucidated, the anti-apoptotic function of A20 is largely unknown. Here, we report a novel mechanism underlying the anti-apoptotic function of A20: A20 blocks TNF-induced apoptosis through suppression of c-jun N-terminal kinase (JNK) by targeting apoptosis signal-regulating kinase1 (ASK1). First, the ectopic expression of A20 drastically inhibits TNF-induced JNK activation and apoptosis in multiple cell types including those deficient of NF-κB activation. Unexpectedly, the blunting effect of A20 on TNF-induced JNK activation is not mediated by affecting the TNFR1 signaling complex formation. Instead, A20 interacts with ASK1, an important MAPKK kinase in the JNK signaling cascade. More importantly, overexpression of wild-type A20, but not of mutant A20 (ZnF4; C624A, C627A), promotes degradation of the ASK1 through the ubiquitin-proteasome system. Taken together, the results from this study reveal a novel anti-apoptotic mechanism of A20 in TNF signaling pathway: A20 binds to ASK1 and mediates ASK1 degradation, leading to suppression of JNK activation and eventually blockage of apoptosis.
Collapse
Affiliation(s)
- M Won
- Department of Pharmacology, Research Institute for Medical Science, Infection Signaling Network Research Center, Daejeon Regional Cancer Center, College of Medicine, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Flygare JA, Fairbrother WJ. Small-molecule pan-IAP antagonists: a patent review. Expert Opin Ther Pat 2010; 20:251-67. [PMID: 20100005 DOI: 10.1517/13543770903567077] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The inhibitor of apoptosis (IAP) proteins are critical regulators of cancer cell survival that have become important targets for therapeutic intervention in human malignancies. One strategy for targeting IAP proteins involves agents that mimic the amino terminus of the endogenous IAP protein antagonist second mitochondria-derived activator of caspases (Smac)/direct IAP-binding protein with low pI (DIABLO) and thus block critical IAP protein interactions. AREAS COVERED IN THIS REVIEW This review of the IAP antagonist patent literature covers the period from 2000 to mid-2009. Over 50 patents and patent applications pertaining to IAP antagonists have been published over the past 10 years. In the case of several filings, only the original source is reviewed in this analysis. WHAT THE READER WILL GAIN Readers will gain an overview of IAP protein antagonist scaffolds, with representative examples including monovalent and bivalent Smac mimetics, and an understanding of their structure-activity relationships. TAKE HOME MESSAGE The feasibility of disrupting IAP protein interactions with pro-apoptotic proteins using monovalent and bivalent Smac-derived peptidomimetic compounds has been broadly established. Four such compounds have entered or been approved to enter human clinical trials, which will hopefully allow the utility of this potential therapeutic approach to be evaluated in cancer patients.
Collapse
Affiliation(s)
- John A Flygare
- Genentech, Inc., Department of Medicinal Chemistry, 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
48
|
Nagai H, Noguchi T, Homma K, Katagiri K, Takeda K, Matsuzawa A, Ichijo H. Ubiquitin-like sequence in ASK1 plays critical roles in the recognition and stabilization by USP9X and oxidative stress-induced cell death. Mol Cell 2010; 36:805-18. [PMID: 20005844 DOI: 10.1016/j.molcel.2009.10.016] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 06/05/2009] [Accepted: 09/04/2009] [Indexed: 11/26/2022]
Abstract
Ubiquitination is an important posttranslational modification that regulates various cellular processes, including signal transduction. However, physiological roles of ubiquitination in the regulation of MAPK pathways are poorly understood. Here, we identified the deubiquitinating enzyme USP9X as a binding partner of ASK1 that mediates oxidative stress-induced cell death through activation of the JNK and p38 MAPK pathways. In the recognition of ubiquitin by deubiquitinating enzymes, the importance of a tandem glycine-glycine sequence in the ubiquitin C terminus has been suggested. Interestingly, ASK1 contains six amino acids identical to the ubiquitin C terminus (LRLRGG), and the GG sequence of ASK1 was required for the USP9X-ASK1 interaction. We also found that USP9X interacted with oxidative stress-activated ASK1 and prevented it from undergoing ubiquitin-dependent degradation. In USP9X-deficient cells, oxidative stress-induced JNK activation and subsequent cell death were reduced. These results demonstrate that USP9X-dependent stabilization of activated ASK1 plays a crucial role in oxidative stress-induced cell death.
Collapse
Affiliation(s)
- Hiroaki Nagai
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, Strategic Approach to Drug Discovery and Development in Pharmaceutical Sciences, Global Center of Excellence Program and Core Research for Evolutional Science and Technology, Japan Science and Technology Corporation, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
Silke J, Brink R. Regulation of TNFRSF and innate immune signalling complexes by TRAFs and cIAPs. Cell Death Differ 2010; 17:35-45. [PMID: 19680262 DOI: 10.1038/cdd.2009.114] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
There have been a number of recent discoveries relating to the functions of inhibitors of apoptosis (IAPs) and TNF receptor-associated factors (TRAFs) in regulating signalling from TNF receptor superfamily (TNFRSF) members and some tantalizing glimpses into a wider area of influence, that of innate immune signalling. Discoveries relating to the function of these ubiquitin E3 ligases in regulating signalling from the eponymous member of the family, TNF-R1, are dealt with superbly in a separate review by Wertz and Dixit and so we will confine our discussion to the subset of the TNFRSF that does not contain a death domain (DD). In line with the available data we will divide the review into two parts, the first is restricted to the role of TRAFs 2 and 3 and cIAPs in regulating TNFRSF signalling, whereas the second will be more speculative, asking what role IAPs and TRAFs have in innate immune signalling.
Collapse
Affiliation(s)
- J Silke
- Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia.
| | | |
Collapse
|
50
|
Vince JE, Pantaki D, Feltham R, Mace PD, Cordier SM, Schmukle AC, Davidson AJ, Callus BA, Wong WWL, Gentle IE, Carter H, Lee EF, Walczak H, Day CL, Vaux DL, Silke J. TRAF2 must bind to cellular inhibitors of apoptosis for tumor necrosis factor (tnf) to efficiently activate nf-{kappa}b and to prevent tnf-induced apoptosis. J Biol Chem 2009; 284:35906-15. [PMID: 19815541 PMCID: PMC2791019 DOI: 10.1074/jbc.m109.072256] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor (TNF) receptor-associated factor-2 (TRAF2) binds to cIAP1 and cIAP2 (cIAP1/2) and recruits them to the cytoplasmic domain of several members of the TNF receptor (TNFR) superfamily, including the TNF-TNFR1 ligand-receptor complex. Here, we define a cIAP1/2-interacting motif (CIM) within the TRAF-N domain of TRAF2, and we use TRAF2 CIM mutants to determine the role of TRAF2 and cIAP1/2 individually, and the TRAF2-cIAP1/2 interaction, in TNFR1-dependent signaling. We show that both the TRAF2 RING domain and the TRAF2 CIM are required to regulate NF-kappaB-inducing kinase stability and suppress constitutive noncanonical NF-kappaB activation. Conversely, following TNFR1 stimulation, cells bearing a CIM-mutated TRAF2 showed reduced canonical NF-kappaB activation and TNF-induced RIPK1 ubiquitylation. Remarkably, the RING domain of TRAF2 was dispensable for these functions. However, like the TRAF2 CIM, the RING domain of TRAF2 was required for protection against TNF-induced apoptosis. These results show that TRAF2 has anti-apoptotic signaling roles in addition to promoting NF-kappaB signaling and that efficient activation of NF-kappaB by TNFR1 requires the recruitment of cIAP1/2 by TRAF2.
Collapse
Affiliation(s)
- James E. Vince
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Delara Pantaki
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Rebecca Feltham
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Peter D. Mace
- the Biochemistry Department, University of Otago, Dunedin 9054, New Zealand
| | - Stephanie M. Cordier
- the Department of Immunology, Tumour Immunology Unit, Division of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom, and
| | - Anna C. Schmukle
- the Department of Immunology, Tumour Immunology Unit, Division of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom, and
| | - Angelina J. Davidson
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Bernard A. Callus
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Wendy Wei-Lynn Wong
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Ian E. Gentle
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Holly Carter
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - Erinna F. Lee
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Henning Walczak
- the Department of Immunology, Tumour Immunology Unit, Division of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom, and
| | - Catherine L. Day
- the Biochemistry Department, University of Otago, Dunedin 9054, New Zealand
| | - David L. Vaux
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| | - John Silke
- From the Department of Biochemistry, La Trobe University, Kingsbury Drive, Melbourne, Victoria 3086, Australia
| |
Collapse
|