1
|
Cabana VC, Sénécal AM, Bouchard AY, Kourrich S, Cappadocia L, Lussier MP. AP-1 contributes to endosomal targeting of the ubiquitin ligase RNF13 via a secondary and novel non-canonical binding motif. J Cell Sci 2024; 137:jcs262035. [PMID: 39206621 DOI: 10.1242/jcs.262035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cellular trafficking between organelles is typically assured by short motifs that contact carrier proteins to transport them to their destination. The ubiquitin E3 ligase RING finger protein 13 (RNF13), a regulator of proliferation, apoptosis and protein trafficking, localizes to endolysosomal compartments through the binding of a dileucine motif to clathrin adaptor protein complex AP-3. Mutations within this motif reduce the ability of RNF13 to interact with AP-3. Here, our study shows the discovery of a glutamine-based motif that resembles a tyrosine-based motif within the C-terminal region of RNF13 that binds to the clathrin adaptor protein complex AP-1, notably without a functional interaction with AP-3. Using biochemical, molecular and cellular approaches in HeLa cells, our study demonstrates that a RNF13 dileucine variant uses an AP-1-dependent pathway to be exported from the Golgi towards the endosomal compartment. Overall, this study provides mechanistic insights into the alternate route used by this variant of the dileucine sorting motif of RNF13.
Collapse
Affiliation(s)
- Valérie C Cabana
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Audrey M Sénécal
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Antoine Y Bouchard
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Saïd Kourrich
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Laurent Cappadocia
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| | - Marc P Lussier
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada
- Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines (PROTEO), Montréal, QC H3C 3P8, Canada
| |
Collapse
|
2
|
Le Goupil S, Laprade H, Aubry M, Chevet E. Exploring the IRE1 interactome: From canonical signaling functions to unexpected roles. J Biol Chem 2024; 300:107169. [PMID: 38494075 PMCID: PMC11007444 DOI: 10.1016/j.jbc.2024.107169] [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: 11/13/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
The unfolded protein response is a mechanism aiming at restoring endoplasmic reticulum (ER) homeostasis and is likely involved in other adaptive pathways. The unfolded protein response is transduced by three proteins acting as sensors and triggering downstream signaling pathways. Among them, inositol-requiring enzyme 1 alpha (IRE1α) (referred to as IRE1 hereafter), an endoplasmic reticulum-resident type I transmembrane protein, exerts its function through both kinase and endoribonuclease activities, resulting in both X-box binding protein 1 mRNA splicing and RNA degradation (regulated ire1 dependent decay). An increasing number of studies have reported protein-protein interactions as regulators of these signaling mechanisms, and additionally, driving other noncanonical functions. In this review, we deliver evolutive and structural insights on IRE1 and further describe how this protein interaction network (interactome) regulates IRE1 signaling abilities or mediates other cellular processes through catalytic-independent mechanisms. Moreover, we focus on newly discovered targets of IRE1 kinase activity and discuss potentially novel IRE1 functions based on the nature of the interactome, thereby identifying new fields to explore regarding this protein's biological roles.
Collapse
Affiliation(s)
- Simon Le Goupil
- INSERM U1242, University of Rennes, Rennes, France; Centre de Lutte contre le cancer Eugène Marquis, Rennes, France.
| | - Hadrien Laprade
- INSERM U1242, University of Rennes, Rennes, France; Centre de Lutte contre le cancer Eugène Marquis, Rennes, France
| | - Marc Aubry
- INSERM U1242, University of Rennes, Rennes, France; Centre de Lutte contre le cancer Eugène Marquis, Rennes, France
| | - Eric Chevet
- INSERM U1242, University of Rennes, Rennes, France; Centre de Lutte contre le cancer Eugène Marquis, Rennes, France
| |
Collapse
|
3
|
Clarke CJ, Snider AJ. Perspective: Therapeutic Implications for Sphingolipids in Health and Disease. Mol Pharmacol 2024; 105:118-120. [PMID: 38360837 DOI: 10.1124/molpharm.124.000866] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
Long thought to be structural components of cell membranes, sphingolipids (SLs) have emerged as bioactive molecules whose metabolism is tightly regulated. These bioactive lipids and their metabolic enzymes have been implicated in numerous disease states, including lysosomal storage disorders, multiple sclerosis, inflammation, and cancer as well as metabolic syndrome and obesity. In addition, the indications for many of these lipids to potentially serve as biomarkers for disease continue to emerge with increasing metabolomic and lipidomic studies. The implications of these studies have, in turn, led to the examination of SL enzymes and their bioactive lipids as potential therapeutic targets and as markers for therapeutic efficacy. SIGNIFICANCE STATEMENT: Many sphingolipids (SLs) and their metabolizing enzymes have been implicated in disease. This perspective highlights the potential for SLs to serve as therapeutic targets and diagnostic markers and discusses the implications for the studies and reviews highlighted in this Special Section on Therapeutic Implications for Sphingolipids in Health and Disease.
Collapse
Affiliation(s)
- Christopher J Clarke
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, New York (C.J.C.) and Nutritional Sciences and Wellness, College of Agriculture, Life and Environmental Sciences, University of Arizona Cancer Center, University of Arizona, Tucson, Arizona (A.J.S.)
| | - Ashley J Snider
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, New York (C.J.C.) and Nutritional Sciences and Wellness, College of Agriculture, Life and Environmental Sciences, University of Arizona Cancer Center, University of Arizona, Tucson, Arizona (A.J.S.)
| |
Collapse
|
4
|
Guo S, Zhang BB, Gao L, Yu XY, Shen JH, Yang F, Zhang WC, Jin YG, Li G, Wang YG, Han ZY, Liu Y. RNF13 protects against pathological cardiac hypertrophy through p62-NRF2 pathway. Free Radic Biol Med 2023; 209:252-264. [PMID: 37852547 DOI: 10.1016/j.freeradbiomed.2023.10.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Heart failure (HF) severely impairs human health because of its high incidence and mortality. Cardiac hypertrophy is the main cause of HF, while its underlying mechanism is not fully clear. As an E3 ubiquitin ligase, Ring finger protein 13 (RNF13) plays a crucial role in many disorders, such as liver immune, neurological disease and tumorigenesis, whereas the function of RNF13 in cardiac hypertrophy remains largely unknown. In the present study, we found that the protein expression of RNF13 is up-regulated in the transverse aortic constriction (TAC)-induced murine hypertrophic hearts and phenylephrine (PE)-induced cardiomyocyte hypertrophy. Functional investigations indicated that RNF13 global knockout mice accelerates the degree of TAC-induced cardiac hypertrophy, including cardiomyocyte enlargement, cardiac fibrosis and heart dysfunction. On the contrary, adeno-associated virus 9 (AAV9) mediated-RNF13 overexpression mice alleviated cardiac hypertrophy. Furthermore, we demonstrated that adenoviral RNF13 attenuates the PE-induced cardiomyocyte hypertrophy and down-regulates the expression of cardiac hypertrophic markers, while the opposite results were observed in the RNF13 knockdown group. The RNA-sequence of RNF13 knockout and wild type mice showed that RNF13 deficiency activates oxidative stress after TAC surgery. In terms of the mechanism, we found that RNF13 directly interacted with p62 and promoted the activation of downstream NRF2/HO-1 signaling. Finally, we proved that p62 knockdown can reverse the effect of RNF13 in cardiac hypertrophy. In conclusion, RNF13 protects against the cardiac hypertrophy via p62-NRF2 axis.
Collapse
Affiliation(s)
- Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| | - Bin-Bin Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Xiao-Yue Yu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Ji-Hong Shen
- Department of Electrocardiogram, The Second Affiliated Hospital of Zhengzhou University, No.2 Jingba Road, Zhengzhou, China
| | - Fan Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Wen-Cai Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Ya-Ge Jin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Gang Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Yan-Ge Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China
| | - Zhan-Ying Han
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| | - Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| |
Collapse
|
5
|
Christianson JC, Jarosch E, Sommer T. Mechanisms of substrate processing during ER-associated protein degradation. Nat Rev Mol Cell Biol 2023; 24:777-796. [PMID: 37528230 DOI: 10.1038/s41580-023-00633-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 08/03/2023]
Abstract
Maintaining proteome integrity is essential for long-term viability of all organisms and is overseen by intrinsic quality control mechanisms. The secretory pathway of eukaryotes poses a challenge for such quality assurance as proteins destined for secretion enter the endoplasmic reticulum (ER) and become spatially segregated from the cytosolic machinery responsible for disposal of aberrant (misfolded or otherwise damaged) or superfluous polypeptides. The elegant solution provided by evolution is ER-membrane-bound ubiquitylation machinery that recognizes misfolded or surplus proteins or by-products of protein biosynthesis in the ER and delivers them to 26S proteasomes for degradation. ER-associated protein degradation (ERAD) collectively describes this specialized arm of protein quality control via the ubiquitin-proteasome system. But, instead of providing a single strategy to remove defective or unwanted proteins, ERAD represents a collection of independent processes that exhibit distinct yet overlapping selectivity for a wide range of substrates. Not surprisingly, ER-membrane-embedded ubiquitin ligases (ER-E3s) act as central hubs for each of these separate ERAD disposal routes. In these processes, ER-E3s cooperate with a plethora of specialized factors, coordinating recognition, transport and ubiquitylation of undesirable secretory, membrane and cytoplasmic proteins. In this Review, we focus on substrate processing during ERAD, highlighting common threads as well as differences between the many routes via ERAD.
Collapse
Affiliation(s)
- John C Christianson
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| | - Ernst Jarosch
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany
| | - Thomas Sommer
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany.
- Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Lin Z, Yang P, Hu Y, Xu H, Duan J, He F, Dou K, Wang L. RING finger protein 13 protects against nonalcoholic steatohepatitis by targeting STING-relayed signaling pathways. Nat Commun 2023; 14:6635. [PMID: 37857628 PMCID: PMC10587083 DOI: 10.1038/s41467-023-42420-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disorder worldwide. Recent studies show that innate immunity-related signaling pathways fuel NAFLD progression. This study aims to identify potent regulators of innate immunity during NAFLD progression. To this end, a phenotype-based high-content screening is performed, and RING finger protein 13 (RNF13) is identified as an effective inhibitor of lipid accumulation in vitro. In vivo gain- and loss-of-function assays are conducted to investigate the role of RNF13 in NAFLD. Transcriptome sequencing and immunoprecipitation-mass spectrometry are performed to explore the underlying mechanisms. We reveal that RNF13 protein is upregulated in the liver of individuals with NASH. Rnf13 knockout in hepatocytes exacerbate insulin resistance, steatosis, inflammation, cell injury and fibrosis in the liver of diet-induced mice, which can be alleviated by Rnf13 overexpression. Mechanically, RNF13 facilitates the proteasomal degradation of stimulator of interferon genes protein (STING) in a ubiquitination-dependent way. This study provides a promising innate immunity-related target for NAFLD treatment.
Collapse
Affiliation(s)
- Zhibin Lin
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Peijun Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yufeng Hu
- Gannan Innovation and Transformation Medical Research Institute, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, 341000, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fei He
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
7
|
He J, Ding Y, Lin H, Liu X, Chen X, Shen W, Zhou S, Feng C, Wang M, Xia J, He N. Differential genome-wide associated variants and enriched pathways of ECG parameters among people with versus without HIV. AIDS 2023; 37:1871-1882. [PMID: 37418550 PMCID: PMC10481915 DOI: 10.1097/qad.0000000000003647] [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: 03/02/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/09/2023]
Abstract
OBJECTIVES People with HIV (PWH) are more likely to develop ECG abnormalities. Substantial evidence exists for genetic contribution to ECG parameters among general population. However, whether and how would host genome associate with ECG parameters among PWH is unclear. Our research aims to analyze and compare genetic variants, mapped genes, and enriched pathways of ECG parameters among PWH and HIV-negative controls. DESIGN A cross-sectional study. METHOD We performed a large original genome-wide association study (GWAS) of ECG parameters among PWH ( n = 1730) and HIV-negative controls ( n = 3746). Genome-wide interaction analyses were also conducted. RESULTS A total of 18 novel variants were detected among PWH, six for PR interval including rs76345397 at ATL2 , 11 for QRS duration including rs10483994 at KCNK10 and rs2478830 at JCAD , and one for QTc interval (rs9815364). Among HIV-negative controls, we identified variants located at previously reported ECG-related genes ( SCN5A , CNOT1 ). Genetic variants had a significant interaction with HIV infection ( P < 5 × 10 -8 ), implying that HIV infection and host genome might jointly influence ECG parameters. Mapped genes for PR interval and QRS duration among PWH were enriched in the biological process of viral genome replication and host response to virus, respectively, whereas enriched pathways for PR interval among HIV-negative controls were in the cellular component of voltage-gated sodium channel complex. CONCLUSION The present GWAS indicated a distinctive impact of host genome on quantitative ECG parameters among PWH. Different from HIV-negative controls, host genome might influence the cardiac electrical activity by interfering with HIV viral infection, production, and latency among PWH.
Collapse
Affiliation(s)
- Jiayu He
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| | - Yingying Ding
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Haijiang Lin
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Xing Liu
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Xiaoxiao Chen
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Weiwei Shen
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Sujuan Zhou
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| | - Cheng Feng
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Miaochen Wang
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Jingjing Xia
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Na He
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| |
Collapse
|
8
|
Bankier S, Wang L, Crawford A, Morgan RA, Ruusalepp A, Andrew R, Björkegren JLM, Walker BR, Michoel T. Plasma cortisol-linked gene networks in hepatic and adipose tissues implicate corticosteroid-binding globulin in modulating tissue glucocorticoid action and cardiovascular risk. Front Endocrinol (Lausanne) 2023; 14:1186252. [PMID: 37745713 PMCID: PMC10513085 DOI: 10.3389/fendo.2023.1186252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/14/2023] [Indexed: 09/26/2023] Open
Abstract
Genome-wide association meta-analysis (GWAMA) by the Cortisol Network (CORNET) consortium identified genetic variants spanning the SERPINA6/SERPINA1 locus on chromosome 14 associated with morning plasma cortisol, cardiovascular disease (CVD), and SERPINA6 mRNA expression encoding corticosteroid-binding globulin (CBG) in the liver. These and other findings indicate that higher plasma cortisol levels are causally associated with CVD; however, the mechanisms by which variations in CBG lead to CVD are undetermined. Using genomic and transcriptomic data from The Stockholm Tartu Atherosclerosis Reverse Networks Engineering Task (STARNET) study, we identified plasma cortisol-linked single-nucleotide polymorphisms (SNPs) that are trans-associated with genes from seven different vascular and metabolic tissues, finding the highest representation of trans-genes in the liver, subcutaneous fat, and visceral abdominal fat, [false discovery rate (FDR) = 15%]. We identified a subset of cortisol-associated trans-genes that are putatively regulated by the glucocorticoid receptor (GR), the primary transcription factor activated by cortisol. Using causal inference, we identified GR-regulated trans-genes that are responsible for the regulation of tissue-specific gene networks. Cis-expression Quantitative Trait Loci (eQTLs) were used as genetic instruments for identification of pairwise causal relationships from which gene networks could be reconstructed. Gene networks were identified in the liver, subcutaneous fat, and visceral abdominal fat, including a high confidence gene network specific to subcutaneous adipose (FDR = 10%) under the regulation of the interferon regulatory transcription factor, IRF2. These data identify a plausible pathway through which variation in the liver CBG production perturbs cortisol-regulated gene networks in peripheral tissues and thereby promote CVD.
Collapse
Affiliation(s)
- Sean Bankier
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
- Division of Genetics and Genomics, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Lingfei Wang
- Division of Genetics and Genomics, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Crawford
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Ruth A. Morgan
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- SRUC, The Roslin Institute, Edinburgh, United Kingdom
| | - Arno Ruusalepp
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia
- Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
- Clinical Gene Networks AB, Stockholm, Sweden
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Johan L. M. Björkegren
- Clinical Gene Networks AB, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Brian R. Walker
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tom Michoel
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
- Division of Genetics and Genomics, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Kline G, Paxman RJ, Lin CY, Madrazo N, Yoon L, Grandjean JMD, Lee K, Nugroho K, Powers ET, Wiseman RL, Kelly JW. Divergent Proteome Reactivity Influences Arm-Selective Activation of the Unfolded Protein Response by Pharmacological Endoplasmic Reticulum Proteostasis Regulators. ACS Chem Biol 2023; 18:1719-1729. [PMID: 37523656 PMCID: PMC10442855 DOI: 10.1021/acschembio.3c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/05/2023] [Indexed: 08/02/2023]
Abstract
Pharmacological activation of the activating transcription factor 6 (ATF6) arm of the unfolded protein response (UPR) has proven useful for ameliorating proteostasis deficiencies in cellular and mouse models of numerous etiologically diverse diseases. Previous high-throughput screening efforts identified the small molecule AA147 as a potent and selective ATF6 activating compound that operates through a mechanism involving metabolic activation of its 2-amino-p-cresol substructure affording a quinone methide, which then covalently modifies a subset of endoplasmic reticulum (ER) protein disulfide isomerases (PDIs). Another compound identified in this screen, AA132, also contains a 2-amino-p-cresol moiety; however, this compound showed less transcriptional selectivity, instead globally activating all three arms of the UPR. Here, we show that AA132 activates global UPR signaling through a mechanism analogous to that of AA147, involving metabolic activation and covalent modification of proteins including multiple PDIs. Chemoproteomic-enabled analyses show that AA132 covalently modifies PDIs to a greater extent than AA147. However, the extent of PDI labeling by AA147 approaches a plateau more rapidly than PDI labeling by AA132. These observations together suggest that AA132 can access a larger pool of proteins for covalent modification, possibly because its activated form is less susceptible to quenching than activated AA147. In other words, the lower reactivity of activated AA132 allows it to persist longer and modify more PDIs in the cellular environment. Collectively, these results suggest that AA132 globally activates the UPR through increased engagement of ER PDIs. Consistent with this, reducing the cellular concentration of AA132 decreases PDI modifications and enables selective ATF6 activation. Our results highlight the relationship between metabolically activatable-electrophile stability, ER proteome reactivity, and the transcriptional response observed with the enaminone chemotype of ER proteostasis regulators, enabling continued development of next-generation ATF6 activating compounds.
Collapse
Affiliation(s)
- Gabriel
M. Kline
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Ryan J. Paxman
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Chung-Yon Lin
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Nicole Madrazo
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Leonard Yoon
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Julia M. D. Grandjean
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Kyunga Lee
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Karina Nugroho
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Evan T. Powers
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - R. Luke Wiseman
- Department
of Molecular Medicine, The Scripps Research
Institute, La Jolla, California 92037, United States
| | - Jeffery W. Kelly
- Department
of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
- The
Skaggs Institute for Chemical Biology, The
Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
10
|
Kline GM, Paxman RJ, Lin CY, Madrazo N, Grandjean JM, Lee K, Nugroho K, Powers ET, Wiseman RL, Kelly JW. Divergent Proteome Reactivity Influences Arm-Selective Activation of Pharmacological Endoplasmic Reticulum Proteostasis Regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524237. [PMID: 36712115 PMCID: PMC9882204 DOI: 10.1101/2023.01.16.524237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Pharmacological activation of the activating transcription factor 6 (ATF6) arm of the Unfolded Protein Response (UPR) has proven useful for ameliorating proteostasis deficiencies in a variety of etiologically diverse diseases. Previous high-throughput screening efforts identified the small molecule AA147 as a potent and selective ATF6 activating compound that operates through a mechanism involving metabolic activation of its 2-amino- p -cresol substructure affording a quinone methide, which then covalently modifies a subset of ER protein disulfide isomerases (PDIs). Intriguingly, another compound identified in this screen, AA132, also contains a 2-amino- p -cresol moiety; however, this compound showed less transcriptional selectivity, instead globally activating all three arms of the UPR. Here, we show that AA132 activates global UPR signaling through a mechanism analogous to that of AA147, involving metabolic activation and covalent PDI modification. Chemoproteomic-enabled analyses show that AA132 covalently modifies PDIs to a greater extent than AA147. Paradoxically, activated AA132 reacts slower with PDIs, indicating it is less reactive than activated AA147. This suggests that the higher labeling of PDIs observed with activated AA132 can be attributed to its lower reactivity, which allows this activated compound to persist longer in the cellular environment prior to quenching by endogenous nucleophiles. Collectively, these results suggest that AA132 globally activates the UPR through increased engagement of ER PDIs. Consistent with this, reducing the cellular concentration of AA132 decreases PDI modifications and allows for selective ATF6 activation. Our results highlight the relationship between metabolically activatable-electrophile stability, ER proteome reactivity, and the transcriptional response observed with the enaminone chemotype of ER proteostasis regulators, enabling continued development of next-generation ATF6 activating compounds.
Collapse
Affiliation(s)
- Gabriel M. Kline
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Ryan J Paxman
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Chung-Yon Lin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Nicole Madrazo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Julia M. Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Kyunga Lee
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Karina Nugroho
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - Evan T. Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
11
|
Fu F, Doroudgar S. IRE1/XBP1 and endoplasmic reticulum signaling - from basic to translational research for cardiovascular disease. CURRENT OPINION IN PHYSIOLOGY 2022; 28:100552. [PMID: 37207249 PMCID: PMC10195104 DOI: 10.1016/j.cophys.2022.100552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Most cellular protein synthesis, including synthesis of membrane-targeted and secreted proteins, which are critical for cellular and organ crosstalk, takes place at the endoplasmic reticulum (ER), placing the ER at the nexus of cellular signaling, growth, metabolism, and stress sensing. Ample evidence has established the dysregulation of protein homeostasis and the ER unfolded protein response (UPR) in cardiovascular disease. However, the mechanisms of stress sensing and signaling in the ER are incompletely defined. Recent studies have defined notable functions for the inositol-requiring kinase 1 (IRE1)/X-box- binding protein-1 (XBP1) branch of the UPR in regulation of cardiac function. This review highlights the mechanisms underlying IRE1 activation and the IRE1 interactome, which reveals unexpected functions for the UPR and summarizes our current understanding of the functions of IRE1 in cardiovascular disease.
Collapse
Affiliation(s)
- Fangyi Fu
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Shirin Doroudgar
- Department of Internal Medicine and the Translational Cardiovascular Research Center, University of Arizona - College of Medicine - Phoenix, Phoenix, AZ, United States
| |
Collapse
|
12
|
Khani M, Nafissi S, Shamshiri H, Moazzeni H, Taheri H, Elahi E. Identification of RNF13 as cause of recessively inherited ALS in a multi-case pedigree. J Med Genet 2022; 60:jmedgenet-2022-108645. [PMID: 35879052 DOI: 10.1136/jmg-2022-108645] [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: 04/18/2022] [Accepted: 07/05/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease. The approximately 50 known ALS-associated genes do not fully explain its heritability, which suggests the existence of yet unidentified causative genes. We report results of studies aimed at identification of the genetic cause of ALS in a pedigree (three patients) without mutations in the common ALS-causative genes. METHODS Clinical investigations included thorough neurological and non-neurological examinations and testings. Genetic analysis was performed by exome sequencing. Functional studies included identification of altered splicing by PCR and sequencing, and mutated proteins by western blot analysis. Apoptosis in the presence and absence of tunicamycin was assessed in transfected HEK293T cells using an Annexin-PE-7AAD kit in conjunction with flow cytometry. RESULTS Clinical features are described in detail. Disease progression in the patients of the pedigree was relatively slow and survival was relatively long. An RNF13 mutation was identified as the cause of the recessively inherited ALS in the pedigree. The gene is highly conserved, and its encoded protein (RING finger protein 13) can potentially affect various neurodegenerative-relevant functions, including protein homeostasis. The RNF13 splice site mutation caused expression of two mis-spliced forms of RNF13 mRNA and an aberrant RNF13 protein, and affected apoptosis. CONCLUSION RNF13 was identified as a novel causative gene of recessively inherited ALS. The gene affects protein homeostasis, which is one of most important components of the pathology of neurodegeneration. The contribution of RNF13 to the aetiology of another neurodegenerative disease is discussed.
Collapse
Affiliation(s)
- Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Shahriar Nafissi
- Department of Neurology, Tehran University of Medical Sciences, Tehran, The Islamic Republic of Iran
| | - Hosein Shamshiri
- Department of Neurology, Tehran University of Medical Sciences, Tehran, The Islamic Republic of Iran
| | - Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Hanieh Taheri
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, The Islamic Republic of Iran
| |
Collapse
|
13
|
Banerjee DK, Seijo Lebrón A, Baksi K. Glycotherapy: A New Paradigm in Breast Cancer Research. Biomolecules 2022; 12:biom12040487. [PMID: 35454076 PMCID: PMC9026886 DOI: 10.3390/biom12040487] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is an ancient disease recognized first by the Egyptians as early as 1600 BC. The first cancer-causing gene in a chicken tumor virus was found in 1970. The United States signed the National Cancer Act in 1971, authorizing federal funding for cancer research. Irrespective of multi-disciplinary approaches, diverting a great deal of public and private resources, breast cancer remains at the forefront of human diseases, affecting as many as one in eight women during their lifetime. Because of overarching challenges and changes in the breast cancer landscape, five-year disease-free survival is no longer considered adequate. The absence of a cure, and the presence of drug resistance, severe side effects, and destruction of the patient’s quality of life, as well as the fact that therapy is often expensive, making it unaffordable to many, have created anxiety among patients, families, and friends. One of the reasons for the failure of cancer therapeutics is that the approaches do not consider cancer holistically. Characteristically, all breast cancer cells and their microenvironmental capillary endothelial cells express asparagine-linked (N-linked) glycoproteins with diverse structures. We tested a small biological molecule, Tunicamycin, that blocks a specific step of the protein N-glycosylation pathway in the endoplasmic reticulum (ER), i.e., the catalytic activity of N-acetylglusosaminyl 1-phosphate transferase (GPT). The outcome was overwhelmingly exciting. Tunicamycin quantitatively inhibits angiogenesis in vitro and in vivo, and inhibits the breast tumor progression of multiple subtypes in pre-clinical mouse models with “zero” toxicity. Mechanistic details support ER stress-induced unfolded protein response (upr) signaling as the cause for the apoptotic death of both cancer and the microvascular endothelial cells. Additionally, it interferes with Wnt signaling. We therefore conclude that Tunicamycin can be expected to supersede the current therapeutics to become a glycotherapy for treating breast cancer of all subtypes.
Collapse
Affiliation(s)
- Dipak K. Banerjee
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936-5067, USA;
- Correspondence:
| | - Arelis Seijo Lebrón
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936-5067, USA;
| | - Krishna Baksi
- Department of Anatomy and Cell Biology, School of Medicine, Universidad Central del Caribe, Bayamon, PR 00960-3001, USA;
| |
Collapse
|
14
|
From Drosophila to Human: Biological Function of E3 Ligase Godzilla and Its Role in Disease. Cells 2022; 11:cells11030380. [PMID: 35159190 PMCID: PMC8834447 DOI: 10.3390/cells11030380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin–proteasome system is of fundamental importance in all fields of biology due to its impact on proteostasis and in regulating cellular processes. Ubiquitination, a type of protein post-translational modification, involves complex enzymatic machinery, such as E3 ubiquitin ligases. The E3 ligases regulate the covalent attachment of ubiquitin to a target protein and are involved in various cellular mechanisms, including the cell cycle, cell division, endoplasmic reticulum stress, and neurotransmission. Because the E3 ligases regulate so many physiological events, they are also associated with pathologic conditions, such as cancer, neurological disorders, and immune-related diseases. This review focuses specifically on the protease-associated transmembrane-containing the Really Interesting New Gene (RING) subset of E3 ligases. We describe the structure, partners, and physiological functions of the Drosophila Godzilla E3 ligase and its human homologues, RNF13, RNF167, and ZNRF4. Also, we summarize the information that has emerged during the last decade regarding the association of these E3 ligases with pathophysiological conditions, such as cancer, asthma, and rare genetic disorders. We conclude by highlighting the limitations of the current knowledge and pinpointing the unresolved questions relevant to RNF13, RNF167, and ZNRF4 ubiquitin ligases.
Collapse
|
15
|
Zhou Z, Wang Q, Michalak M. Inositol Requiring Enzyme (IRE), a multiplayer in sensing endoplasmic reticulum stress. Anim Cells Syst (Seoul) 2022; 25:347-357. [PMID: 35059134 PMCID: PMC8765250 DOI: 10.1080/19768354.2021.2020901] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Zhixin Zhou
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
Cabana VC, Bouchard AY, Sénécal AM, Ghilarducci K, Kourrich S, Cappadocia L, Lussier MP. RNF13 Dileucine Motif Variants L311S and L312P Interfere with Endosomal Localization and AP-3 Complex Association. Cells 2021; 10:cells10113063. [PMID: 34831286 PMCID: PMC8620429 DOI: 10.3390/cells10113063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEE) are rare and serious neurological disorders characterized by severe epilepsy with refractory seizures and a significant developmental delay. Recently, DEE73 was linked to genetic alterations of the RNF13 gene, which convert positions 311 or 312 in the RNF13 protein from leucine to serine or proline, respectively (L311S and L312P). Using a fluorescence microscopy approach to investigate the molecular and cellular mechanisms affected by RNF13 protein variants, the current study shows that wild-type RNF13 localizes extensively with endosomes and lysosomes, while L311S and L312P do not extensively colocalize with the lysosomal marker Lamp1. Our results show that RNF13 L311S and L312P proteins affect the size of endosomal vesicles along with the temporal and spatial progression of fluorescently labeled epidermal growth factor, but not transferrin, in the endolysosomal system. Furthermore, GST-pulldown and co-immunoprecipitation show that RNF13 variants disrupt association with AP-3 complex. Knockdown of AP-3 complex subunit AP3D1 alters the lysosomal localization of wild-type RNF13 and similarly affects the size of endosomal vesicles. Importantly, our study provides a first step toward understanding the cellular and molecular mechanism altered by DEE73-associated genetic variations of RNF13.
Collapse
Affiliation(s)
- Valérie C. Cabana
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Antoine Y. Bouchard
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Audrey M. Sénécal
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Kim Ghilarducci
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Saïd Kourrich
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laurent Cappadocia
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
| | - Marc P. Lussier
- Département de Chimie, Université du Québec à Montréal, Montréal, QC H2X 2J6, Canada; (V.C.C.); (A.Y.B.); (A.M.S.); (K.G.); (L.C.)
- Centre d’Excellence en Recherche sur les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montréal, QC H2X 3Y7, Canada;
- Correspondence: ; Tel.: +1-(514)-987-3000 (ext. 5591); Fax: +1-(514)-987-4054
| |
Collapse
|
17
|
MiR-32-3p Regulates Myocardial Injury Induced by Microembolism and Microvascular Obstruction by Targeting RNF13 to Regulate the Stability of Atherosclerotic Plaques. J Cardiovasc Transl Res 2021; 15:143-166. [PMID: 34185281 DOI: 10.1007/s12265-021-10150-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
This study aimed to explore the molecular mechanism of myocardial protection. The effects of miR-32-3p and ring finger protein 13 (RNF13) on endoplasmic reticulum (ER) stress-induced apoptosis of A-10 cells and human umbilical vein endothelial cells (HUVEC) were detected using flow cytometry. The effects of miR-32-3p and phenylbutyric acid (PBA) on plaque instability and myocardial tissue injury in rats were investigated after establishment of arterial plaque model and embolization model and treatment with miR-32-3p-antagomir and PBA. RNF13, which was differentially expressed in myocardial infarction, was the direct target gene of miR-32-3p. MiR-32-3p inhibited RNF13 expression and targeted RNF13 to inhibit ER stress-induced cell apoptosis. Furthermore, inhibiting miR-32-3p expression induced arterial plaque instability by reducing survival, increasing pathological lesions in arterial tissue, up-regulating ER stress-related proteins, and regulating the expressions of apoptosis-related proteins in the model rats. However, PBA reversed the effects of miR-32-3p-antagomir on the model rats. MiR-32-3p regulates myocardial injury induced by micro-embolism and micro-vascular obstruction by targeting RNF13 to regulate the stability of atherosclerotic plaques.
Collapse
|
18
|
Reticular Basement Membrane Thickness Is Associated with Growth- and Fibrosis-Promoting Airway Transcriptome Profile-Study in Asthma Patients. Int J Mol Sci 2021; 22:ijms22030998. [PMID: 33498209 PMCID: PMC7863966 DOI: 10.3390/ijms22030998] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/24/2022] Open
Abstract
Airway remodeling in asthma is characterized by reticular basement membrane (RBM) thickening, likely related to epithelial structural and functional changes. Gene expression profiling of the airway epithelium might identify genes involved in bronchial structural alterations. We analyzed bronchial wall geometry (computed tomography (CT)), RBM thickness (histology), and the bronchial epithelium transcriptome profile (gene expression array) in moderate to severe persistent (n = 21) vs. no persistent (n = 19) airflow limitation asthmatics. RBM thickness was similar in the two studied subgroups. Among the genes associated with increased RBM thickness, the most essential were those engaged in cell activation, proliferation, and growth (e.g., CDK20, TACC2, ORC5, and NEK5) and inhibiting apoptosis (e.g., higher mRNA expression of RFN34, BIRC3, NAA16, and lower of RNF13, MRPL37, CACNA1G). Additionally, RBM thickness correlated with the expression of genes encoding extracellular matrix (ECM) components (LAMA3, USH2A), involved in ECM remodeling (LTBP1), neovascularization (FGD5, HPRT1), nerve functioning (TPH1, PCDHGC4), oxidative stress adaptation (RIT1, HSP90AB1), epigenetic modifications (OLMALINC, DNMT3A), and the innate immune response (STAP1, OAS2). Cluster analysis revealed that genes linked with RBM thickness were also related to thicker bronchial walls in CT. Our study suggests that the pro-fibrotic profile in the airway epithelial cell transcriptome is associated with a thicker RBM, and thus, may contribute to asthma airway remodeling.
Collapse
|
19
|
Ness C, Katta K, Garred Ø, Kumar T, Olstad OK, Petrovski G, Moe MC, Noer A. Integrated differential DNA methylation and gene expression of formalin-fixed paraffin-embedded uveal melanoma specimens identifies genes associated with early metastasis and poor prognosis. Exp Eye Res 2020; 203:108426. [PMID: 33387485 DOI: 10.1016/j.exer.2020.108426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE Uveal melanoma (UM) is an aggressive malignancy, in which nearly 50% of the patients die from metastatic disease. Aberrant DNA methylation is recognized as an important epigenomic event in carcinogenesis. Formalin-fixed paraffin-embedded (FFPE) samples represent a valuable source of tumor tissue, and recent technology has enabled the use of these samples in genome-wide DNA methylation analyses. Our aim was to investigate differential DNA methylation in relation to histopathological classification and survival data. In addition we sought to identify aberrant DNA methylation of genes that could be associated with metastatic disease and poor survival. METHODS FFPE samples from UM patients (n = 23) who underwent enucleation of the eye in the period 1976-1989 were included. DNA methylation was assessed using the Illumina Infinium HumanMethylation450 array and coupled to histopathological data, Cancer Registry of Norway- (registered UM metastasis) and Norwegian Cause of Death Registry- (time and cause of death) data. Differential DNA methylation patterns contrasting histological classification, survival data and clustering properties were investigated. Survival groups were defined as "Early metastasis" (metastases and death within 2-5 years after enucleation, n = 8), "Late metastasis" (metastases and death within 9-21 years after enucleation, n = 7) and "No metastasis" (no detected metastases ≥18 years after enucleation, n = 8). A subset of samples were selected based on preliminary multi-dimensional scaling (MDS) plots, histopathological classification, chromosome 3 status, survival status and clustering properties; "Subset Early metastasis" (n = 4) vs "Subset No metastasis" (n = 4). Bioinformatics analyses were conducted in the R statistical software. Differentially methylated positions (DMPs) and differentially methylated regions (DMRs) in various comparisons were assessed. Gene expression of relevant subgroups was determined by microarray analysis and quantitative reverse-transcription polymerase chain reaction (qRT-PCR). RESULTS DNA methylation analyses identified 2 clusters that separated the samples according to chromosome 3 status. Cluster 1 consisted of samples (n = 5) with chromosome 3 disomy (D3), while Cluster 2 was comprised of samples (n = 15) with chromosome 3 monosomy (M3). 1212 DMRs and 9386 DMPs were identified in M3 vs D3. No clear clusters were formed based on our predefined survival groups ("Early", "Late", "No") nor histopathological classification (Epithelioid, Mixed, Spindle). We identified significant changes in DNA methylation (beta FC ≥ 0.2, adjusted p < 0.05) between two sample subsets (n = 8). "Subset Early metastasis" (n = 4) vs "Subset No metastasis" (n = 4) identified 348 DMPs and 36 DMRs, and their differential gene expression by microarray showed that 14 DMPs and 2 DMRs corresponded to changes in gene expression (FC ≥ 1.5, p < 0.05). RNF13, ZNF217 and HYAL1 were hypermethylated and downregulated in "Subset Early metastasis" vs "Subset No metastasis" and could be potential tumor suppressors. TMEM200C, RGS10, ADAM12 and PAM were hypomethylated and upregulated in "Subset Early metastasis vs "Subset No metastasis" and could be potential oncogenes and thus markers of early metastasis and poor prognosis in UM. CONCLUSIONS DNA methylation profiling showed differential clustering of samples according to chromosome 3 status: Cluster 1 (D3) and Cluster 2 (M3). Integrated differential DNA methylation and gene expression of two subsets of samples identified genes associated with early metastasis and poor prognosis. RNF13, ZNF217 and HYAL1 are hypermethylated and candidate tumor suppressors, while TMEM200C, RGS10, ADAM12 and PAM are hypomethylated and candidate oncogenes linked to early metastasis. UM FFPE samples represent a valuable source for methylome studies and enable long-time follow-up.
Collapse
Affiliation(s)
- Charlotte Ness
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kirankumar Katta
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Norway
| | - Theresa Kumar
- Department of Pathology, Oslo University Hospital, Norway
| | | | - Goran Petrovski
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Morten C Moe
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Agate Noer
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
20
|
Bashir S, Banday M, Qadri O, Bashir A, Hilal N, Nida-I-Fatima, Rader S, Fazili KM. The molecular mechanism and functional diversity of UPR signaling sensor IRE1. Life Sci 2020; 265:118740. [PMID: 33188833 DOI: 10.1016/j.lfs.2020.118740] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum is primarily responsible for protein folding and maturation. However, the organelle is subject to varied stress conditions from time to time, which lead to the activation of a signaling program known as the Unfolded Protein Response (UPR) pathway. This pathway, upon sensing any disturbance in the protein-folding milieu sends signals to the nucleus and cytoplasm in order to restore homeostasis. One of the prime UPR signaling sensors is Inositol-requiring enzyme 1 (IRE1); an ER membrane embedded protein with dual enzyme activities, kinase and endoribonuclease. The ribonuclease activity of IRE1 results in Xbp1 splicing in mammals or Hac1 splicing in yeast. However, IRE1 can switch its substrate specificity to the mRNAs that are co-transnationally transported to the ER, a phenomenon known as Regulated IRE1 Dependent Decay (RIDD). IRE1 is also reported to act as a principal molecule that coordinates with other proteins and signaling pathways, which in turn might be responsible for its regulation. The current review highlights studies on IRE1 explaining the structural features and molecular mechanism behind its ribonuclease outputs. The emphasis is also laid on the molecular effectors, which directly or indirectly interact with IRE1 to either modulate its function or connect it to other pathways. This is important in understanding the functional pleiotropy of IRE1, by which it can switch its activity from pro-survival to pro-apoptotic, thus determining the fate of cells.
Collapse
Affiliation(s)
- Samirul Bashir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Mariam Banday
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Ozaira Qadri
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Arif Bashir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Nazia Hilal
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Nida-I-Fatima
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Stephen Rader
- Department of Chemistry, University of Northern British Columbia, Prince George, BC, Canada
| | - Khalid Majid Fazili
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India.
| |
Collapse
|
21
|
Urra H, Pihán P, Hetz C. The UPRosome - decoding novel biological outputs of IRE1α function. J Cell Sci 2020; 133:133/15/jcs218107. [PMID: 32788208 DOI: 10.1242/jcs.218107] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Different perturbations alter the function of the endoplasmic reticulum (ER), resulting in the accumulation of misfolded proteins in its lumen, a condition termed ER stress. To restore ER proteostasis, a highly conserved pathway is engaged, known as the unfolded protein response (UPR), triggering adaptive programs or apoptosis of terminally damaged cells. IRE1α (also known as ERN1), the most conserved UPR sensor, mediates the activation of responses to determine cell fate under ER stress. The complexity of IRE1α regulation and its signaling outputs is mediated in part by the assembly of a dynamic multi-protein complex, named the UPRosome, that regulates IRE1α activity and the crosstalk with other pathways. We discuss several studies identifying components of the UPRosome that have illuminated novel functions in cell death, autophagy, DNA damage, energy metabolism and cytoskeleton dynamics. Here, we provide a theoretical analysis to assess the biological significance of the UPRosome and present the results of a systematic bioinformatics analysis of the available IRE1α interactome data sets followed by functional enrichment clustering. This in silico approach decoded that IRE1α also interacts with proteins involved in the cell cycle, transport, differentiation, response to viral infection and immune response. Thus, defining the spectrum of IRE1α-binding partners will reveal novel signaling outputs and the relevance of the pathway to human diseases.
Collapse
Affiliation(s)
- Hery Urra
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile .,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile
| | - Philippe Pihán
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8380453, Chile .,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago 8380453, Chile.,The Buck Institute for Research in Aging, Novato, CA 94945, USA
| |
Collapse
|
22
|
Role of RING-Type E3 Ubiquitin Ligases in Inflammatory Signalling and Inflammatory Bowel Disease. Mediators Inflamm 2020; 2020:5310180. [PMID: 32848509 PMCID: PMC7436281 DOI: 10.1155/2020/5310180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/22/2020] [Indexed: 01/05/2023] Open
Abstract
Ubiquitination is a three-step enzymatic cascade for posttranslational protein modification. It includes the ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2), and ubiquitin ligase (E3). RING-type E3 ubiquitin ligases catalyse the posttranslational proteolytic and nonproteolytic functions in various physiological and pathological processes, such as inflammation-associated signal transduction. Resulting from the diversity of substrates and functional mechanisms, RING-type ligases regulate microbe recognition and inflammation by being involved in multiple inflammatory signalling pathways. These processes also occur in autoimmune diseases, especially inflammatory bowel disease (IBD). To understand the importance of RING-type ligases in inflammation, we have discussed their functional mechanisms in multiple inflammation-associated pathways and correlation between RING-type ligases and IBD. Owing to the limited data on the biology of RING-type ligases, there is an urgent need to analyse their potential as biomarkers and therapeutic targets in IBD in the future.
Collapse
|
23
|
Ji M, Niu S, Guo J, Mi H, Jiang P. Silencing RNF13 Alleviates Parkinson’s Disease – Like Problems in Mouse Models by Regulating the Endoplasmic Reticulum Stress–Mediated IRE1α-TRAF2-ASK1-JNK Pathway. J Mol Neurosci 2020; 70:1977-1986. [DOI: 10.1007/s12031-020-01599-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022]
|
24
|
Riaz TA, Junjappa RP, Handigund M, Ferdous J, Kim HR, Chae HJ. Role of Endoplasmic Reticulum Stress Sensor IRE1α in Cellular Physiology, Calcium, ROS Signaling, and Metaflammation. Cells 2020; 9:E1160. [PMID: 32397116 PMCID: PMC7290600 DOI: 10.3390/cells9051160] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) is the most prominent and evolutionarily conserved unfolded protein response (UPR) signal transducer during endoplasmic reticulum functional upset (ER stress). A IRE1α signal pathway arbitrates yin and yang of cellular fate in objectionable conditions. It plays several roles in fundamental cellular physiology as well as in several pathological conditions such as diabetes, obesity, inflammation, cancer, neurodegeneration, and in many other diseases. Thus, further understanding of its molecular structure and mechanism of action during different cell insults helps in designing and developing better therapeutic strategies for the above-mentioned chronic diseases. In this review, recent insights into structure and mechanism of activation of IRE1α along with its complex regulating network were discussed in relation to their basic cellular physiological function. Addressing different binding partners that can modulate IRE1α function, UPRosome triggers different downstream pathways depending on the cellular backdrop. Furthermore, IRE1α are in normal cell activities outside the dominion of ER stress and activities under the weather of inflammation, diabetes, and obesity-related metaflammation. Thus, IRE1 as an ER stress sensor needs to be understood from a wider perspective for comprehensive functional meaning, which facilitates us with assembling future needs and therapeutic benefits.
Collapse
Affiliation(s)
- Thoufiqul Alam Riaz
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Mallikarjun Handigund
- Department of Laboratory Medicine, Jeonbuk National University, Medical School, Jeonju 54907, Korea;
| | - Jannatul Ferdous
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Hyung-Ryong Kim
- College of Dentistry, Dankook University, Cheonan 31116, Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| |
Collapse
|
25
|
Andrawus M, Sharvit L, Shekhidem HA, Roichman A, Cohen HY, Atzmon G. The effects of environmental stressors on candidate aging associated genes. Exp Gerontol 2020; 137:110952. [PMID: 32344118 DOI: 10.1016/j.exger.2020.110952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/29/2020] [Accepted: 04/09/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Aging is defined as a biological and physical complex process that is characterized by the increase in susceptibility to diseases and eventually death. Aging may occur at different rates between and within species, especially or (it varies) among the long-lived ones. Here, we ask whether this diversity (e.g. aging phenotype) stems from genetic or environmental factors or as a combination between the two (epigenetics). Epigenetics play a central role in controlling changes in gene expression during aging. DNA methylation is the most abundant epigenetic modification among vertebrates and is essential to mammalian development. MATERIALS AND METHODS In this study, we utilized the HELPtag assay to identify five candidate genes that were significantly hyper- or hypo-methylated across four different age groups in mice. The candidate genes were annotated using ensemble and their expression was further tested in vitro using the murine RAW 264.7 cell line to examine the effect of three environmental stressors (UV radiation, Hypoxia and fasting) on their expression. RNA was extracted at different time points followed by cDNA synthesis. Changes in gene expression were evaluated using qRT-PCR. RESULTS We show that fasting and UV radiation reduced the viability of RAW264.7 cells. We also found a significant change in three candidate genes' expression levels during fasting (TOP2B, RNF13 and MRPL4). Furthermore, we found a significant change in the four candidate genes' expression levels following UVC treatment (TOP2B, RNF13, PKNOX1 and CREB5) and yet no changes were recorded in hypoxic conditions. CONCLUSION Our results suggest that the model we used was a fitting model for the assessment of environmental stressors on candidate gene expression. In addition, we established a cellular response to the environment via changes in gene expression.
Collapse
Affiliation(s)
- Mariana Andrawus
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel
| | - Lital Sharvit
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel
| | | | - Asael Roichman
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Haim Y Cohen
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Gil Atzmon
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
26
|
Cen S, Li J, Cai Z, Pan Y, Sun Z, Li Z, Ye G, Zheng G, Li M, Liu W, Yu W, Wang S, Xie Z, Wang P, Shen H. TRAF4 acts as a fate checkpoint to regulate the adipogenic differentiation of MSCs by activating PKM2. EBioMedicine 2020; 54:102722. [PMID: 32268273 PMCID: PMC7191261 DOI: 10.1016/j.ebiom.2020.102722] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) selectively differentiate into adipocytes or osteoblasts, and several molecules control the fate determination of MSCs. Understanding these key checkpoints greatly contributes to the ability to induce specific MSC differentiation for clinical applications. In this study, we aimed to explore whether TNF receptor-associated factor 4 (TRAF4) affects MSC adipogenic differentiation, which we previously reported that could positively regulated the osteogenic differentiation. METHODS Western blotting and Real-time Polymerase Chain Reaction were used to detected the expression pattern of TRAF4 during adipogenic differentiation. Lentivirus was constructed to regulate TRAF4 expression, and oil red O staining and Western blotting were used to assess its role in adipogenesis, which was confirmed in vivo by implanting an MSC-matrigel mixture into nude mice. Western blotting was used to detect the activated signaling pathways, and a specific inhibitor and agonist were used to clear the roles of the key signaling pathways. Additionaly, Co-Immunoprecipitation was conducted to find that Pyruvate kinase isozyme type M2 (PKM2) interacts with TRAF4, and to further explore their binding and functional domains. Finally, an RNA-binding protein immunoprecipitation assay and Western blotting were used to detect whether N6-methyladenosine mediates the decreased TRAF4 expression during adipogenic differentiation. FINDINGS The results demonstrated that TRAF4 negatively regulates MSC adipogenesis in vitro and in vivo. Mechanistically, we revealed that TRAF4 binds to PKM2 to activate the kinase activity of PKM2, which subsequently activates β-catenin signaling and then inhibits adipogenesis. Furthermore, TRAF4 downregulation during adipogenesis is regulated by ALKBH5-mediated N6-methyladenosine RNA demethylation. INTERPRETATION TRAF4 negatively regulates the adipogenesis of MSCs by activating PKM2 kinase activity, which may act as a checkpoint to fine-tune the balance of adipo-osteogenic differentiation, and suggests that TRAF4 may be a novel target of MSCs in clinical use and may also illuminate the underlying mechanisms of bone metabolic diseases. FUNDING This study was supported by the National Natural Science Foundation of China (81871750 and 81971518) and the Science and Technology Project of Guangdong Province (2019B02023600 and 2017A020215070).
Collapse
Affiliation(s)
- Shuizhong Cen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China
| | - Yiqian Pan
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Zehang Sun
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Zhaofeng Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Guiwen Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China
| | - Ming Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Wenhui Yu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China
| | - Shan Wang
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518003, PR China; Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, PR China.
| |
Collapse
|
27
|
Lindner P, Christensen SB, Nissen P, Møller JV, Engedal N. Cell death induced by the ER stressor thapsigargin involves death receptor 5, a non-autophagic function of MAP1LC3B, and distinct contributions from unfolded protein response components. Cell Commun Signal 2020; 18:12. [PMID: 31987044 PMCID: PMC6986015 DOI: 10.1186/s12964-019-0499-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cell death triggered by unmitigated endoplasmic reticulum (ER) stress plays an important role in physiology and disease, but the death-inducing signaling mechanisms are incompletely understood. To gain more insight into these mechanisms, the ER stressor thapsigargin (Tg) is an instrumental experimental tool. Additionally, Tg forms the basis for analog prodrugs designed for cell killing in targeted cancer therapy. Tg induces apoptosis via the unfolded protein response (UPR), but how apoptosis is initiated, and how individual effects of the various UPR components are integrated, is unclear. Furthermore, the role of autophagy and autophagy-related (ATG) proteins remains elusive. METHODS To systematically address these key questions, we analyzed the effects of Tg and therapeutically relevant Tg analogs in two human cancer cell lines of different origin (LNCaP prostate- and HCT116 colon cancer cells), using RNAi and inhibitory drugs to target death receptors, UPR components and ATG proteins, in combination with measurements of cell death by fluorescence imaging and propidium iodide staining, as well as real-time RT-PCR and western blotting to monitor caspase activity, expression of ATG proteins, UPR components, and downstream ER stress signaling. RESULTS In both cell lines, Tg-induced cell death depended on death receptor 5 and caspase-8. Optimal cytotoxicity involved a non-autophagic function of MAP1LC3B upstream of procaspase-8 cleavage. PERK, ATF4 and CHOP were required for Tg-induced cell death, but surprisingly acted in parallel rather than as a linear pathway; ATF4 and CHOP were independently required for Tg-mediated upregulation of death receptor 5 and MAP1LC3B proteins, whereas PERK acted via other pathways. Interestingly, IRE1 contributed to Tg-induced cell death in a cell type-specific manner. This was linked to an XBP1-dependent activation of c-Jun N-terminal kinase, which was pro-apoptotic in LNCaP but not HCT116 cells. Molecular requirements for cell death induction by therapy-relevant Tg analogs were identical to those observed with Tg. CONCLUSIONS Together, our results provide a new, integrated understanding of UPR signaling mechanisms and downstream mediators that induce cell death upon Tg-triggered, unmitigated ER stress. Video Abstract.
Collapse
Affiliation(s)
- Paula Lindner
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership for Molecular Medicine, University of Oslo, P.O. Box 1137, Blindern, N-0318 Oslo, Norway
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Poul Nissen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Nikolai Engedal
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership for Molecular Medicine, University of Oslo, P.O. Box 1137, Blindern, N-0318 Oslo, Norway
| |
Collapse
|
28
|
Moura J, Madureira P, Leal EC, Fonseca AC, Carvalho E. Immune aging in diabetes and its implications in wound healing. Clin Immunol 2019; 200:43-54. [PMID: 30735729 PMCID: PMC7322932 DOI: 10.1016/j.clim.2019.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Immune systems have evolved to recognize and eliminate pathogens and damaged cells. In humans, it is estimated to recognize 109 epitopes and natural selection ensures that clonally expanded cells replace unstimulated cells and overall immune cell numbers remain stationary. But, with age, it faces continuous repertoire restriction and concomitant accumulation of primed cells. Changes shaping the aging immune system have bitter consequences because, as inflammatory responses gain intensity and duration, tissue-damaging immunity and inflammatory disease arise. During inflammation, the glycolytic flux cannot cope with increasing ATP demands, limiting the immune response's extent. In diabetes, higher glucose availability stretches the glycolytic limit, dysregulating proteostasis and increasing T-cell expansion. Long-term hyperglycemia exerts an accumulating effect, leading to higher inflammatory cytokine levels and increased cytotoxic mediator secretion upon infection, a phenomenon known as diabetic chronic inflammation. Here we review the etiology of diabetic chronic inflammation and its consequences on wound healing.
Collapse
Affiliation(s)
- J Moura
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| | - P Madureira
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, University of Porto, Porto, Portugal; Immunethep, Biocant Park, Cantanhede, Portugal
| | - E C Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - A C Fonseca
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - E Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Coimbra, Portugal; Department of Geriatrics, University of Arkansas for Medical Sciences and Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
29
|
Edvardson S, Nicolae CM, Noh GJ, Burton JE, Punzi G, Shaag A, Bischetsrieder J, De Grassi A, Pierri CL, Elpeleg O, Moldovan GL. Heterozygous RNF13 Gain-of-Function Variants Are Associated with Congenital Microcephaly, Epileptic Encephalopathy, Blindness, and Failure to Thrive. Am J Hum Genet 2019; 104:179-185. [PMID: 30595371 DOI: 10.1016/j.ajhg.2018.11.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/29/2018] [Indexed: 01/14/2023] Open
Abstract
Accumulation of unfolded proteins in the endoplasmic reticulum (ER) initiates a stress response mechanism to clear out the unfolded proteins by either facilitating their re-folding or inducing their degradation. When this fails, an apoptotic cascade is initiated so that the affected cell is eliminated. IRE1α is a critical sensor of the unfolded-protein response, essential for initiating the apoptotic signaling. Here, we report an infantile neurodegenerative disorder associated with enhanced activation of IRE1α and increased apoptosis. Three unrelated affected individuals with congenital microcephaly, infantile epileptic encephalopathy, and profound developmental delay were found to carry heterozygous variants (c.932T>C [p.Leu311Ser] or c.935T>C [p.Leu312Pro]) in RNF13, which codes for an IRE1α-interacting protein. Structural modeling predicted that the variants, located on the surface of the protein, would not alter overall protein folding. Accordingly, the abundance of RNF13 and IRE1α was not altered in affected individuals' cells. However, both IRE1α-mediated stress signaling and stress-induced apoptosis were increased in affected individuals' cells. These results indicate that the RNF13 variants confer gain of function to the encoded protein and thereby lead to altered signaling of the ER stress response associated with severe neurodegeneration in infancy.
Collapse
Affiliation(s)
- Simon Edvardson
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; Pediatric Neurology Unit, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Claudia M Nicolae
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Grace J Noh
- Department of Genetics, Southern California Permanente Medical Group, Fontana, CA 92335, USA
| | - Jennifer E Burton
- University of Illinois College of Medicine at Peoria, Illini Drive, Peoria, IL 61605, USA
| | - Giuseppe Punzi
- Laboratory of Biochemistry, Molecular and Computational Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari 70125, Italy
| | - Avraham Shaag
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Jessica Bischetsrieder
- Department of Genetics, Southern California Permanente Medical Group, Fontana, CA 92335, USA
| | - Anna De Grassi
- Laboratory of Biochemistry, Molecular and Computational Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari 70125, Italy
| | - Ciro Leonardo Pierri
- Laboratory of Biochemistry, Molecular and Computational Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari 70125, Italy
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| | - George-Lucian Moldovan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
30
|
Mike EV, Makinde HM, Gulinello M, Vanarsa K, Herlitz L, Gadhvi G, Winter DR, Mohan C, Hanly JG, Mok CC, Cuda CM, Putterman C. Lipocalin-2 is a pathogenic determinant and biomarker of neuropsychiatric lupus. J Autoimmun 2019; 96:59-73. [PMID: 30174216 PMCID: PMC6310639 DOI: 10.1016/j.jaut.2018.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 11/22/2022]
Abstract
Neuropsychiatric manifestations in lupus (NPSLE) affect ∼20-40% of patients. In the central nervous system, lipocalin-2 (LCN2) can promote injury through mechanisms directly linked to NPSLE, including brain barrier disruption, neurotoxicity, and glial activation. Since LCN2 is elevated in lupus and has been implicated in neuroinflammation, we investigated whether LCN2 is required for the pathogenesis of NPSLE. Here, we investigated the effects of LCN2 deficiency on the development of neurobehavioral deficits in the B6.Sle1.Sle3 (Sle1,3) mouse lupus model. Sle1,3 mice exhibited depression-like behavior and impaired spatial and recognition memory, and these deficits were attenuated in Sle1,3-LCN2KO mice. Whole-brain flow cytometry showed a significant increase in brain infiltrating leukocytes in Sle1,3 mice that was not reduced by LCN2 deficiency. RNA sequencing on sorted microglia revealed that several genes differentially expressed between B6 and Sle1,3 mice were regulated by LCN2, and that these genes are key mediators of the neuroinflammatory cascade. Importantly, LCN2 is upregulated in the cerebrospinal fluid of NPSLE patients across 2 different ethnicities. Our findings establish the Sle1,3 strain as an NPSLE model, demonstrate that LCN2 is a major regulator of the detrimental neuroimmune response in NPSLE, and identify CSF LCN2 as a novel biomarker for NPSLE.
Collapse
Affiliation(s)
- Elise V Mike
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hadijat M Makinde
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Maria Gulinello
- Dominick P. Purpura Department of Neuroscience Animal Behavioral Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Leal Herlitz
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, 44915, USA
| | - Gaurav Gadhvi
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Deborah R Winter
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - John G Hanly
- Division of Rheumatology, Department of Medicine and Department of Pathology, Dalhousie University and Queen Elizabeth II Health Sciences Center, Halifax, Nova Scotia, Canada
| | - C C Mok
- Division of Rheumatology, Tuen Mun Hospital, Hong Kong, China
| | - Carla M Cuda
- Division of Rheumatology, Northwestern University School of Medicine, Chicago, IL, USA
| | - Chaim Putterman
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
31
|
Liu XJ, Wei J, Shang YH, Huang HC, Lao FX. Modulation of AβPP and GSK3β by Endoplasmic Reticulum Stress and Involvement in Alzheimer's Disease. J Alzheimers Dis 2018; 57:1157-1170. [PMID: 28339396 DOI: 10.3233/jad-161111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a dementia disease with neuronal loss and synaptic impairment. This impairment is caused, at least partly, by the generation of two main AD hallmarks, namely the hyperphosphorylated tau protein comprising neurofibrillary tangles and senile plaques containing amyloid-β (Aβ) peptides. The amyloid-β protein precursor (AβPP) and glycogen synthase kinase-3β (GSK3β) are two main proteins associated with AD and are closely correlated with these hallmarks. Recently, both of the proteins were reported to be modulated by endoplasmic reticulum stress (ERS) and are involved in the pathogenesis of AD. The mechanism of ERS plus the modulation of AβPP processing and GSK3β activity by ERS in AD are summarized and explored in this review.
Collapse
Affiliation(s)
- Xin-Jun Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Jun Wei
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| |
Collapse
|
32
|
JNK inhibitor alleviates apoptosis of fetal neural stem cells induced by emulsified isoflurane. Oncotarget 2017; 8:94009-94019. [PMID: 29212205 PMCID: PMC5706851 DOI: 10.18632/oncotarget.21505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 09/13/2017] [Indexed: 11/25/2022] Open
Abstract
Isoflurane can provide both neuroprotection and neurotoxicity in various culture models and in rodent developing brains. Emulsified Isoflurane (EI) is an emulsion formulation of isoflurane, while its underlying molecular mechanism of developemental nerve toxicity largely remains unclear. We hypothesized that EI induced fetal neural stem cells (FNSCs) apoptosis, endoplasmic reticulum (ER) stress and c-Jun N-terminal kinase (JNK) activation. FNSCs were isolated from the cortex of SD rats during 14 days of gestation. The cell viability, cell apoptotic rates and the expression of apoptosis-related protein Caspase3, inositol requiring enzyme 1 (IRE1), poly (adenosine diphosphate-ribose) polymerase (PARP), Bax, Bcl-2, JNK, p-JNK and XBP1 were determined. Specific inhibition was performed by siRNA-targeting of JNK in FNSCs. EI could increase the p-JNK, JNK and caspase3 protein expression, the JNK pathway was activated by EI, and EI-induced apoptosis was blocked by inhibiting JNK pathway with SP600125 or JNK-small interfering RNA (siRNA), EI enhanced the level of IRE1, PARP, Bax/Bcl-2 and XBP1, which led FNSCs to apoptosis and ER stress. Meanwhile, dilatation of the ER lumens in FNSCs treated by EI for 24 h was significant. Green fluorescent protein (GFP) positive cell ratios were significantly decreased by FNSCs transfecting with JNK gene silencing. JNK was efficiently silenced in siRNA-JNK1 group. The results provided in-vitro evidence which supports that the underlying mechanisms of EI-induced apoptosis are the induction of ER stress and sequent JNK activation. Together, these data suggest that JNK inhibiting might be applied for improving therapeutic outcomes in anesthestics-induced neurotoxicity. Highlights: 1. Prolonged treatment with high-dose EI decreased the survival level of FNSCs by inducing apoptosis and inhibiting proliferation via the JNK signaling pathway. 2. EI induced ER stress and sequent JNK activation. 3. JNK inhibiting might be applied for improving therapeutic outcomes in anesthestics-induced neurotoxicity
Collapse
|
33
|
Kim SH, Kim KY, Yu SN, Seo YK, Chun SS, Yu HS, Ahn SC. Silibinin induces mitochondrial NOX4-mediated endoplasmic reticulum stress response and its subsequent apoptosis. BMC Cancer 2016; 16:452. [PMID: 27405931 PMCID: PMC4942927 DOI: 10.1186/s12885-016-2516-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 06/27/2016] [Indexed: 12/02/2022] Open
Abstract
Background Silibinin, a biologically active compound of milk thistle, has chemopreventive effects on cancer cell lines. Recently it was reported that silibinin inhibited tumor growth through activation of the apoptotic signaling pathway. Although various evidences showed multiple signaling pathways of silibinin in apoptosis, there were no reports to address the clear mechanism of ROS-mediated pathway in prostate cancer PC-3 cells. Several studies suggested that reactive oxygen species (ROS) play an important role in various signaling cascades, but the primary source of ROS was currently unclear. Methods The effect of silibinin was investigated on cell growth of prostate cell lines by MTT assay. We examined whether silibinin induced apoptosis through production of ROS using flow cytometry. Expression of apoptosis-, endoplasmic reticulum (ER)-related protein and gene were determined by western blotting and RT-PCR, respectively. Results Results showed that silibinin triggered mitochondrial ROS production through NOX4 expression and finally led to induce apoptosis. In addition, mitochondrial ROS caused ER stress through disruption of Ca2+ homeostasis. Co-treatment of ROS inhibitor reduced the silibinin-induced apoptosis through the inhibition of NOX4 expression, resulting in reduction of both Ca2+ level and ER stress response. Conclusions Taken together, silibinin induced mitochondrial ROS-dependent apoptosis through NOX4, which is associated with disruption of Ca2+ homeostasis and ER stress response. Therefore, the regulation of NOX4, mitochondrial ROS producer, could be a potential target for the treatment of prostate cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2516-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sang-Hun Kim
- Department of Microbiology & Immunology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea
| | - Kwang-Youn Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, 689-798, Republic of Korea
| | - Sun-Nyoung Yu
- Department of Microbiology & Immunology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea.,Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Young-Kyo Seo
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, 689-798, Republic of Korea
| | - Sung-Sik Chun
- Department of Food Science, International University of Korea, Jinju, 660-759, Republic of Korea
| | - Hak-Sun Yu
- Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870, Republic of Korea.,Department of Parasitology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea
| | - Soon-Cheol Ahn
- Department of Microbiology & Immunology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea. .,Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, 626-870, Republic of Korea.
| |
Collapse
|
34
|
Brown M, Strudwick N, Suwara M, Sutcliffe LK, Mihai AD, Ali AA, Watson JN, Schröder M. An initial phase of JNK activation inhibits cell death early in the endoplasmic reticulum stress response. J Cell Sci 2016; 129:2317-2328. [PMID: 27122189 DOI: 10.1242/jcs.179127] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 04/20/2016] [Indexed: 12/21/2022] Open
Abstract
Accumulation of unfolded proteins in the endoplasmic reticulum (ER) activates the unfolded protein response (UPR). In mammalian cells, UPR signals generated by several ER-membrane-resident proteins, including the bifunctional protein kinase endoribonuclease IRE1α, control cell survival and the decision to execute apoptosis. Processing of XBP1 mRNA by the RNase domain of IRE1α promotes survival of ER stress, whereas activation of the mitogen-activated protein kinase JNK family by IRE1α late in the ER stress response promotes apoptosis. Here, we show that activation of JNK in the ER stress response precedes activation of XBP1. This activation of JNK is dependent on IRE1α and TRAF2 and coincides with JNK-dependent induction of expression of several antiapoptotic genes, including cIap1 (also known as Birc2), cIap2 (also known as Birc3), Xiap and Birc6 ER-stressed Jnk1(-/-) Jnk2(-/-) (Mapk8(-/-) Mapk9(-/-)) mouse embryonic fibroblasts (MEFs) display more pronounced mitochondrial permeability transition and increased caspase 3/7 activity compared to wild-type MEFs. Caspase 3/7 activity is also elevated in ER-stressed cIap1(-/-) cIap2(-/-) and Xiap(-/-) MEFs. These observations suggest that JNK-dependent transcriptional induction of several inhibitors of apoptosis contributes to inhibiting apoptosis early in the ER stress response.
Collapse
Affiliation(s)
- Max Brown
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Natalie Strudwick
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Monika Suwara
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Louise K Sutcliffe
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Adina D Mihai
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Ahmed A Ali
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK.,Molecular Biology Department, National Research Centre, Dokki 12311, Cairo, Egypt
| | - Jamie N Watson
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| | - Martin Schröder
- Durham University, School of Biological and Biomedical Sciences, Durham DH1 3LE, United Kingdom.,Biophysical Sciences Institute, Durham University, Durham DH1 3LE, United Kingdom.,North East England Stem Cell Institute (NESCI), Life Bioscience Centre, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 4EP, UK
| |
Collapse
|
35
|
Huang XP, Ding H, Lu JD, Tang YH, Deng BX, Deng CQ. Effects of the Combination of the Main Active Components of Astragalus and Panax notoginseng on Inflammation and Apoptosis of Nerve Cell after Cerebral Ischemia-Reperfusion. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1419-38. [DOI: 10.1142/s0192415x15500809] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Astragalus and Panax notoginseng are commonly used to treat cardio-cerebrovascular diseases in China and are often combined together to promote curative effect. We speculate that the enhancement of the combination on anticerebral ischemia injury may come from the main active components. The purpose of this work was to probe the effects and mechanisms of Astragaloside IV (the active component of Astragalus) combined with Ginsenoside Rg1, Ginsenoside Rb1, and Notoginsenoside R1 (the active components of P. notoginseng) to antagonize ischemia/reperfusion (I/R) injury via inflammation and apoptosis. C57BL/6 mice were randomly divided into sham, model, Astragaloside IV, Ginsenoside Rg1, Ginsenoside Rb1, Notoginsenoside R1, four active components combination, and Edaravone groups. After administration for 3 days, bilateral common carotid arteries (CCA) were occluded with artery clip for 20[Formula: see text]min followed by reperfusion for 24[Formula: see text]h. Our results showed that the survival rate of nerve cell in hippocampal CA1 decreased while the apoptotic rate increased, and the level of caspase-3 protein in brain tissues was elevated, the expressions of TNF-a, IL-1, and ICAM-1 mRNA as well as phosphorylated nuclear factor kappa B (NF-[Formula: see text]B) inhibitor protein [Formula: see text] (p-I[Formula: see text]Ba) in brain tissues were up-regulated, and the nuclear translocation rate of NF-[Formula: see text]B was raised. Additionally, the protein expressions of phosphorylated tyrosine kinase 1 (p-JAK1), phosphorylated signal transducer and activator of transcription-1 (p-STAT1), glucose regulated protein 78 (GRP78), caspase-12, and phosphorylated c-Jun N-terminal kinases 1/2 (p-JNK1/2) in brain tissues were also significantly strengthened after I/R for 24[Formula: see text]h. All drugs could increase neurocyte survival rate in hippocampal CA1, decrease the apoptotic rate, and inhibit caspase-3 protein expression, in contrast, the effects of four active components combination were better than those of active components alone. In addition, Astragaloside IV and Ginsenoside Rg1 could down-regulate the level of TNF-[Formula: see text], and ICAM-1 mRNA, respectively, Notoginsenoside R1 reduced both TNF-[Formula: see text] and ICAM-1 mRNA, and the combination of the 4 effective components had inhibitory effects on the expressions of TNF-[Formula: see text], IL-1[Formula: see text], and ICAM-1 mRNA. Astragaloside IV, Ginsenoside Rg1, Notoginsenoside R1, and 4 effective components combination were able to restrain the phosphorylation of I[Formula: see text]B[Formula: see text], and relieve the nuclear translocation rate of NF-[Formula: see text]B. Moreover, the effects of the combination are greater than those of active components alone. All drugs could suppress the phosphorylation of JAK1 induced by I/R; meanwhile the expression of p-STAT1 exhibited a decrease in Ginsenoside Rg1 and four active components combination groups. The decreases of p-JAK1 and p-STAT1 in the four active components combination group were more obvious than those in active components alone groups. Astragaloside IV, Ginsenoside Rg1, and Notoginsenoside R1 further augmented GRP78 expression caused by I/R, Notoginsenoside R1 attenuated caspase-12 protein expression, Astragaloside IV and Ginsenoside Rg1 lessened the phosphorylation of JNK1/2, and the four active components combination was capable of up-regulating GRP78 protein while down-regulating the expressions of caspase-12 and p-JNK1/2. Similarly, the effects of the four active components combination were greater than those of effective components alone. These suggested that the combination of the main active components of Astragalus and Panax notoginseng could strengthen protective effects on cerebral ischemia injury via anti-apoptosis and anti-inflammation, and the mechanisms might be associated with restraining the activation of NF-[Formula: see text]B and JAK1/STAT1 signal pathways and regulating endoplasmic reticulum stress (ERS) after cerebral ischemia.
Collapse
Affiliation(s)
- Xiao-Ping Huang
- Molecular Pathology Laboratory, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, P.R. China
| | - Huang Ding
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated, Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Changsha 410208, Hunan Province, P.R. China
| | - Jin-Dong Lu
- Key Laboratory of Hunan Universities for Cell biology and Molecular Techniques, Changsha 410208, Hunan Province, P.R. China
| | - Ying-Hong Tang
- Key Laboratory of Hunan Province for Prevention and Treatment of Integrated, Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Changsha 410208, Hunan Province, P.R. China
| | - Bing-Xiang Deng
- Key Laboratory of Hunan Universities for Cell biology and Molecular Techniques, Changsha 410208, Hunan Province, P.R. China
| | - Chang-Qing Deng
- Molecular Pathology Laboratory, Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, P.R. China
| |
Collapse
|
36
|
Troxerutin inhibits 2,2′,4,4′-tetrabromodiphenyl ether (BDE-47)-induced hepatocyte apoptosis by restoring proteasome function. Toxicol Lett 2015; 233:246-57. [DOI: 10.1016/j.toxlet.2015.01.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/30/2014] [Accepted: 01/25/2015] [Indexed: 12/25/2022]
|
37
|
Fu XL, Gao DS. Endoplasmic reticulum proteins quality control and the unfolded protein response: the regulative mechanism of organisms against stress injuries. Biofactors 2014; 40:569-85. [PMID: 25530003 DOI: 10.1002/biof.1194] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/25/2014] [Indexed: 12/21/2022]
Abstract
The endoplasmic reticulum is the cellular compartment in which secretory proteins are synthesized and folded. Perturbations of endoplasmic reticulum homeostasis lead to the accumulation of unfolded proteins. The activation of the unfolded protein response during endoplasmic reticulum stress transmits information about the status of protein folding to the cytosol and nucleus. The unfolded protein response leads to the upregulation of genes encoding endoplasmic reticulum chaperones, attenuation of translation, and initiation of the endoplasmic reticulum quality control system to restore endoplasmic reticulum homeostasis. When the unfolded protein response is insufficient to rebuild the steady state in endoplasmic reticulum, the programmed cell death or apoptosis would be initiated, by triggering cell injuries, even to cell death through apoptosis signals. In this review, we briefly outline research on the chaperones and foldases conserved in eukaryotes and plants, and describe the general principles and mechanisms of the endoplasmic reticulum quality control and the unfolded protein response. We describe the current models for the molecular mechanism of the unfolded protein response in plants, and emphasize the role of inositol requiring enzyme-1-dependent network in the unfolded protein response. Finally, we give a general overview of the directions for future research on the unfolded protein response in plants and its role in the response to environmental stresses.
Collapse
Affiliation(s)
- Xi Ling Fu
- Division of National Research Center for Apple Engineering and Technology, Shandong Agricultural University, Tai'an, Shandong, China; Division of State Key Laboratory of Crop Biology, Shandong Agricultural University, Tai'an, Shandong, China; College of Horticulture Science and Engineering, Shandong Agricultural University, Tai'an, Shandong, China
| | | |
Collapse
|
38
|
Zhu X, Zhang J, Sun H, Jiang C, Dong Y, Shan Q, Su S, Xie Y, Xu N, Lou X, Liu S. Ubiquitination of inositol-requiring enzyme 1 (IRE1) by the E3 ligase CHIP mediates the IRE1/TRAF2/JNK pathway. J Biol Chem 2014; 289:30567-30577. [PMID: 25225294 DOI: 10.1074/jbc.m114.562868] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Deciphering the inositol-requiring enzyme 1 (IRE1) signaling pathway is fundamentally important for understanding the unfolded protein response (UPR). The ubiquitination of proteins residing on the endoplasmic reticulum (ER) membrane has been reported to be involved in the UPR, although the mechanism has yet to be fully elucidated. Using immunoprecipitation and mass spectrometry, IRE1 was identified as a substrate of the E3 ligase CHIP (carboxyl terminus of HSC70-interacting protein) in HEK293 cells under geldanamycin-induced ER stress. Two residues of IRE1, Lys(545) and Lys(828), were targeted for Lys(63)-linked ubiquitination. Moreover, in CHIP knockdown cells, IRE1 phosphorylation and the IRE1-TRAF2 interaction were nearly abolished under ER stress, which may be due to lacking ubiquitination of IRE1 on Lys(545) and Lys(828), respectively. The cellular responses were evaluated, and the data indicated that CHIP-regulated IRE1/TRAF2/JNK signaling antagonized the senescence process. Therefore, our findings suggest that CHIP-mediated ubiquitination of IRE1 contributes to the dynamic regulation of the UPR.
Collapse
Affiliation(s)
- Xu Zhu
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China,; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ju Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiying Sun
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Cuicui Jiang
- Beijing Protein Innovation, Beijing 101318, China, and
| | - Yusheng Dong
- Beijing Protein Innovation, Beijing 101318, China, and
| | - Qiang Shan
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Siyuan Su
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China,; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingying Xie
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China,; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaomin Lou
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China,.
| | - Siqi Liu
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China,; University of Chinese Academy of Sciences, Beijing 100049, China,.
| |
Collapse
|
39
|
Tumour-associated mutations of PA-TM-RING ubiquitin ligases RNF167/RNF13 identify the PA domain as a determinant for endosomal localization. Biochem J 2014; 459:27-36. [DOI: 10.1042/bj20131067] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In the present study, we have identified a role for the PA domain of the RNF13/RNF167 ubiquitin ligases, showing that it is both sufficient and required for endosomal localization. Furthermore, although PA domain point-mutations identified from tumours are ligase active, PA mutant proteins are mislocalized within the cell.
Collapse
|
40
|
Arshad A, Gu X, Arshad M. RNF13 protein regulates endoplasmic reticulum stress induced apoptosis in dopaminergic SH-SY5Y cells by enhancing IRE1α stability. J Recept Signal Transduct Res 2013; 34:119-24. [PMID: 24303962 DOI: 10.3109/10799893.2013.863920] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Any interruption in the folding capacity of endoplasmic reticulum (ER) may result in inducing ER stress which would initiate an adaptive signaling mechanism called Unfolded Protein Response (UPR) in order to restore homeostasis, failing to which would initiate signaling pathway leading to death of the cell. Finding new mediators could help better understand the molecular mechanisms of ER stress-induced apoptosis. Our lab initiated a genetic screen method using retroviral insertion mutation system to look for genes whose inactivation would confer resistance to apoptosis. In our previous findings, Ring finger protein 13 (RNF13) was identified whose down-regulation conferred survival against ER stress-induced apoptosis. Our previous results also showed important role of RNF13 in apoptotic signaling in 293T cells as a result of strong RNF13-IRE1α interaction. In current study, using SH-SY5Y cells, overexpression of RNF13 in apoptosis assays and RT-PCR analysis has shown to induce apoptosis as well as splicing of X-box binding Protein 1 (XBP1) confirming its role in ER stress mediated cell death in this cell line as well. Western blot analysis has revealed that overexpression of both N-terminal as well as C-terminal tagged RNF13 resulted in activation of c-Jun N-terminal kinase (JNK) in SH-SY5Y cells. Our co-immunoprecipitation assays in SH-SY5Y cells also showed a strong interaction of RNF13 with IRE1α. Finally, Cycloheximide chase experiment exhibited that RNF13-IRE1α interaction increased the stability of IRE1α. Altogether, our data suggest that RNF13 may act as an important regulator of IRE1α, mediating ER stress-mediated apoptosis in neuroblastoma SH-SY5Y cells.
Collapse
Affiliation(s)
- Abida Arshad
- School of Life Sciences, Beijing Institute of Technology , Beijing , China
| | | | | |
Collapse
|