1
|
Esteban-Collado J, Fernández-Mañas M, Fernández-Moreno M, Maeso I, Corominas M, Serras F. Reactive oxygen species activate the Drosophila TNF receptor Wengen for damage-induced regeneration. EMBO J 2024; 43:3604-3626. [PMID: 39020149 PMCID: PMC11377715 DOI: 10.1038/s44318-024-00155-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 07/19/2024] Open
Abstract
Tumor necrosis factor receptors (TNFRs) control pleiotropic pro-inflammatory functions that range from apoptosis to cell survival. The ability to trigger a particular function will depend on the upstream cues, association with regulatory complexes, and downstream pathways. In Drosophila melanogaster, two TNFRs have been identified, Wengen (Wgn) and Grindelwald (Grnd). Although several reports associate these receptors with JNK-dependent apoptosis, it has recently been found that Wgn activates a variety of other functions. We demonstrate that Wgn is required for survival by protecting cells from apoptosis. This is mediated by dTRAF1 and results in the activation of p38 MAP kinase. Remarkably, Wgn is required for apoptosis-induced regeneration and is activated by the reactive oxygen species (ROS) produced following apoptosis. This ROS activation is exclusive for Wgn, but not for Grnd, and can occur after knocking down Eiger/TNFα. The extracellular cysteine-rich domain of Grnd is much more divergent than that of Wgn, which is more similar to TNFRs from other animals, including humans. Our results show a novel TNFR function that responds to stressors by ensuring p38-dependent regeneration.
Collapse
Affiliation(s)
- José Esteban-Collado
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Mar Fernández-Mañas
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Manuel Fernández-Moreno
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute for Biodiversity Research (IRBio), Barcelona, Spain
- Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Ignacio Maeso
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute for Biodiversity Research (IRBio), Barcelona, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
2
|
Shen J, Lai W, Li Z, Zhu W, Bai X, Yang Z, Wang Q, Ji J. SDS3 regulates microglial inflammation by modulating the expression of the upstream kinase ASK1 in the p38 MAPK signaling pathway. Inflamm Res 2024; 73:1547-1564. [PMID: 39008037 PMCID: PMC11349808 DOI: 10.1007/s00011-024-01913-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Microglia, the main innate immune cells in the central nervous system, are key drivers of neuroinflammation, which plays a crucial role in the pathogenesis of neurodegenerative diseases. The Sin3/histone deacetylase (HDAC) complex, a highly conserved multiprotein co-repressor complex, primarily performs transcriptional repression via deacetylase activity; however, the function of SDS3, which maintains the integrity of the complex, in microglia remains unclear. METHODS To uncover the regulatory role of the transcriptional co-repressor SDS3 in microglial inflammation, we used chromatin immunoprecipitation to identify SDS3 target genes and combined with transcriptomics and proteomics analysis to explore expression changes in cells following SDS3 knocking down. Subsequently, we validated our findings through experimental assays. RESULTS Our analysis revealed that SDS3 modulates the expression of the upstream kinase ASK1 of the p38 MAPK pathway, thus regulating the activation of signaling pathways and ultimately influencing inflammation. CONCLUSIONS Our findings provide important evidence of the contributions of SDS3 toward microglial inflammation and offer new insights into the regulatory mechanisms of microglial inflammatory responses.
Collapse
Affiliation(s)
- Jian Shen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wenjia Lai
- Division of Nanotechnology Development, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zeyang Li
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Wenyuan Zhu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Xue Bai
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Zihao Yang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Averill-Bates D. Reactive oxygen species and cell signaling. Review. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119573. [PMID: 37949302 DOI: 10.1016/j.bbamcr.2023.119573] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 11/12/2023]
Abstract
Reactive oxygen species (ROS) is a term encompassing a group of highly reactive oxygen-derived molecules. In physiological systems, ROS production exists in concert with antioxidant defenses, which safeguard cells against higher, toxic levels of ROS. Oxidative stress, coined as "oxidative distress", is "a serious imbalance between the generation of ROS and antioxidant defenses in favor of ROS, causing excessive oxidative damage to biomolecules". At physiological levels, ROS are essential for many cellular processes, which is known as "oxidative eustress". Oxidants like hydrogen peroxide (H2O2) activate signaling pathways like mitogen-activated protein kinases (MAPK)s and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt). ROS activate transcription factors like nuclear factor erythroid 2-related factor 2 (Nrf2), hypoxia-inducible factor 1α (HIF-1α), activator protein 1 (AP-1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Redox signaling through H2O2 mainly occurs through reversible oxidation of protein cysteine thiolate residues (RS-) to form sulfenic acids (RSOH). An unresolved question is that the reaction rate of H2O2 with protein thiols is very low. In cells, the reaction of H2O2 with protein thiols is likely to be outcompeted by faster reactions of H2O2 with peroxiredoxins and glutathione peroxidases. A novel mechanism being explored is that H2O2 could react with peroxiredoxins that act as reactive redox sensor proteins, leading to peroxiredoxin-mediated relays. Very few redox signaling pathways have been well characterized. Improved understanding of precise mechanisms by which ROS regulate signaling pathways and the role of cellular sensors, is essential for deciphering their roles in physiological and pathological conditions.
Collapse
Affiliation(s)
- Diana Averill-Bates
- Département des sciences biologiques (Center of Excellence in Orphan Diseases Research - Courtois Foundation (CERMO(FC)), Université du Québec à Montréal, Montréal, Québec, Canada.
| |
Collapse
|
4
|
An Y, Wang X, Guan X, Yuan P, Liu Y, Wei L, Wang F, Qi X. Endoplasmic reticulum stress-mediated cell death in cardiovascular disease. Cell Stress Chaperones 2024; 29:158-174. [PMID: 38295944 PMCID: PMC10939083 DOI: 10.1016/j.cstres.2023.12.003] [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: 10/19/2023] [Revised: 12/25/2023] [Accepted: 12/25/2023] [Indexed: 02/24/2024] Open
Abstract
The endoplasmic reticulum (ER) plays a vital function in maintaining cellular homeostasis. Endoplasmic reticulum stress (ERS) can trigger various modes of cell death by activating the unfolded protein response (UPR) signaling pathway. Cell death plays a crucial role in the occurrence and development of diseases such as cancer, liver diseases, neurological diseases, and cardiovascular diseases. Several cardiovascular diseases including hypertension, atherosclerosis, and heart failure are associated with ER stress. ER stress-mediated cell death is of interest in cardiovascular disease. Moreover, an increasing body of evidence supports the potential of modulating ERS for treating cardiovascular disease. This paper provides a comprehensive review of the UPR signaling pathway, the mechanisms that induce cell death, and the modes of cell death in cardiovascular diseases. Additionally, we discuss the mechanisms of ERS and UPR in common cardiovascular diseases, along with potential therapeutic strategies.
Collapse
Affiliation(s)
- Yajuan An
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinshuang Wang
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiuju Guan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng Yuan
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Liu
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
| | - Liping Wei
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
| | - Fei Wang
- Department of Vascular Surgery, Hebei General Hospital, Hebei, China
| | - Xin Qi
- School of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Department of Cardiology, Tianjin Union Medical Center, Tianjin, China.
| |
Collapse
|
5
|
Ogbodo UC, Balogun TA, Omoboyede V. Integrated computational approach identifies potential inhibitors of ASK1-(JNK/P38) interaction signaling: new insights into cancer therapeutics. J Biomol Struct Dyn 2024; 42:696-709. [PMID: 37021478 DOI: 10.1080/07391102.2023.2196699] [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: 01/06/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
Cancers are characterized by the aberrant expression of certain genes that trigger a cascade of molecular events that culminate in dysregulated cell division. Consequently, the inhibition of the products of these expressedgenes has emerged as a rational approach in cancer therapy. The apoptosis signal-regulating kinase 1 (ASK1) protein, encoded by the mitogen-activated protein kinase kinase kinase 5 (MAP3K5) gene, plays pertinent roles in the mediation of cell death induced by stress and inflammation, andis often found at elevated levels in cancer. Consequently, it has emerged as a molecular target for the development of potential chemotherapeutics through identification of selective inhibitors. However, there is still dearth of ASK1 inhibitors in clinical use. Hence, molecular modelling approaches were employed in this study to discover potential ASK1 inhibitors from phytochemicals. Twenty-five phytocompounds from four medicinal plants were tested for their inhibitory prowess via molecular docking. Interestingly, all the compounds exhibited promising inhibitory potentials for ASK1. However, further subjection to filtering procedures via different pipelines including drug-likeness evaluation, pharmacokinetics screening, toxicity profiling, and better affinities compared to the approved inhibitor resulted in three hit compounds namely ellagic acid, luteolin, and kaempferol with suitable properties. Profiling of the interactions formed between the hit\compounds and the targets revealed several interactions that were not present in that of the approved inhibitor, while molecular dynamics (MD) simulation revealed the complexes formed as stable. Conclusively, this study identified three compounds with ASK1 inhibitory potentials that are worthy of further exploration in in vitro and in vivo studies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Uchechukwu C Ogbodo
- Department of Applied Biochemistry, Faculty of Biosciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Toheeb A Balogun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Victor Omoboyede
- Department of Biochemistry, School of Life Sciences (SLS), Federal University of Technology Akure, Akure, Ondo State, Nigeria
- Computer-Aided Therapeutics Laboratory (CATL), School of Life Sciences (SLS), Federal University of Technology Akure, Akure, Nigeria
| |
Collapse
|
6
|
Kostyak JC, McKenzie SE, Naik UP. The Function of ASK1 in Sepsis and Stress-Induced Disorders. Int J Mol Sci 2023; 25:213. [PMID: 38203381 PMCID: PMC10778746 DOI: 10.3390/ijms25010213] [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: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a serine-threonine kinase that is ubiquitously expressed in nucleated cells and is responsible for the activation of multiple mitogen-activated protein kinases (MAPK) to regulate cell stress. Activation of ASK1 via cellular stress leads to activation of downstream signaling components, activation of transcription factors, and proinflammatory cytokine production. ASK1 is also expressed in anucleate platelets and is a key player in platelet activation as it is important for signaling. Interestingly, the mechanism of ASK1 activation is cell type-dependent. In this review we will explore how ASK1 regulates a variety of cellular processes from innate immune function to thrombosis and hemostasis. We will discuss how ASK1 influences FcγRIIA-mediated platelet reactivity and how that reactivity drives platelet clearance. Furthermore, we will explore the role of ASK1 in thromboxane (TxA2) generation, which highlights differences in the way ASK1 functions in mouse and human platelets.
Collapse
Affiliation(s)
- John C. Kostyak
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (S.E.M.); (U.P.N.)
| | | | | |
Collapse
|
7
|
Wang T, Pang L, He M, Wang Z. Small-molecule inhibitors targeting apoptosis signal-regulated kinase 1. Eur J Med Chem 2023; 262:115889. [PMID: 37883895 DOI: 10.1016/j.ejmech.2023.115889] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
Apoptosis signal regulated kinase 1 (ASK1, also known as MAP3K5) is a member of the mitogen activated protein kinase kinase kinase (MAP3K) family. Since its first isolation from a human macrophage library in 1996, its research has been ongoing for over 25 years. A large number of reports have revealed that ASK1, as a key activator of the p38 mitogen-activated protein kinase and c-Jun N-terminal kinase (JNK) signaling cascade, responds to various stressors, and its inhibitors have important potential value in the treatment of diseases such as inflammation, cancer, and the nervous system and so on. This review summarizes the recent development in this field, including the structure and signaling pathways of ASK1, with a particular focus on the structure-activity relationships, and the hit-to-lead optimization strategies.
Collapse
Affiliation(s)
- Tiantian Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Lidan Pang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China
| | - Mengni He
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China
| | - Zengtao Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| |
Collapse
|
8
|
Boyd RA, Majumder S, Stiban J, Mavodza G, Straus AJ, Kempelingaiah SK, Reddy V, Hannun YA, Obeid LM, Senkal CE. The heat shock protein Hsp27 controls mitochondrial function by modulating ceramide generation. Cell Rep 2023; 42:113081. [PMID: 37689067 PMCID: PMC10591768 DOI: 10.1016/j.celrep.2023.113081] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/24/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023] Open
Abstract
Sphingolipids have key functions in membrane structure and cellular signaling. Ceramide is the central molecule of the sphingolipid metabolism and is generated by ceramide synthases (CerS) in the de novo pathway. Despite their critical function, mechanisms regulating CerS remain largely unknown. Using an unbiased proteomics approach, we find that the small heat shock protein 27 (Hsp27) interacts specifically with CerS1 but not other CerS. Functionally, our data show that Hsp27 acts as an endogenous inhibitor of CerS1. Wild-type Hsp27, but not a mutant deficient in CerS1 binding, inhibits CerS1 activity. Additionally, silencing of Hsp27 enhances CerS1-generated ceramide accumulation in cells. Moreover, phosphorylation of Hsp27 modulates Hsp27-CerS1 interaction and CerS1 activity in acute stress-response conditions. Biologically, we show that Hsp27 knockdown impedes mitochondrial function and induces lethal mitophagy in a CerS1-dependent manner. Overall, we identify an important mode of CerS1 regulation and CerS1-mediated mitophagy through protein-protein interaction with Hsp27.
Collapse
Affiliation(s)
- Rowan A Boyd
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Saurav Majumder
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Johnny Stiban
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA; Department of Biology and Biochemistry, Birzeit University, Ramallah, Palestine
| | - Grace Mavodza
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Alexandra J Straus
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Sachin K Kempelingaiah
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA
| | - Varun Reddy
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA; Northport Veterans Affairs Medical Center, Northport, NY 11768, USA
| | - Can E Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23398, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23398, USA.
| |
Collapse
|
9
|
Liu S, Fang X, Zhu R, Zhang J, Wang H, Lei J, Wang C, Wang L, Zhan L. Role of endoplasmic reticulum autophagy in acute lung injury. Front Immunol 2023; 14:1152336. [PMID: 37266445 PMCID: PMC10231642 DOI: 10.3389/fimmu.2023.1152336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), the prime causes of morbidity and mortality in critically ill patients, are usually treated by general supportive treatments. Endoplasmic reticulum autophagy (ER-phagy) maintains cellular homeostasis by degrading damaged endoplasmic reticulum (ER) fragments and misfolded proteins. ER-phagy is crucial for maintaining ER homeostasis and improving the internal environment. ER-phagy has a particular role in some aspects, such as immunity, inflammation, cell death, pathogen infection, and collagen quality. In this review, we summarized the definition, epidemiology, and pathophysiology of ALI/ARDS and described the regulatory mechanisms and functions of ER-phagy as well as discussed the potential role of ER-phagy in ALI/ARDS from the perspectives of immunity, inflammation, apoptosis, pathogen infection, and fibrosis to provide a novel and effective target for improving the prognosis of ALI/ARDS.
Collapse
Affiliation(s)
- Shiping Liu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoyu Fang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruiyao Zhu
- Department of Infection Prevention and Control, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Zhang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huijuan Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiaxi Lei
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chaoqun Wang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Xu N, Liu J, Li X. Therapeutic role of mesenchymal stem cells (MSCs) in diabetic kidney disease (DKD). Endocr J 2022; 69:1159-1172. [PMID: 35858781 DOI: 10.1507/endocrj.ej22-0123] [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] [Indexed: 11/23/2022] Open
Abstract
Findings of preclinical studies and recent phase I/II clinical trials have shown that mesenchymal stem cells (MSCs) play a significant role in the development of diabetic kidney disease (DKD). Thus, MSCs have attracted increasing attention as a novel regenerative therapy for kidney diseases. This review summarizes recent literature on the roles and potential mechanisms, including hyperglycemia regulation, anti-inflammation, anti-fibrosis, pro-angiogenesis, and renal function protection, of MSC-based treatment methods for DKD. This review provides novel insights into understanding the pathogenesis of DKD and guiding the development of biological therapies.
Collapse
Affiliation(s)
- Ning Xu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, 261053, China
| | - Jie Liu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, 261053, China
| | - Xiangling Li
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, 261031, China
| |
Collapse
|
11
|
Cheng Y, Chen J, Shi Y, Fang X, Tang Z. MAPK Signaling Pathway in Oral Squamous Cell Carcinoma: Biological Function and Targeted Therapy. Cancers (Basel) 2022; 14:cancers14194625. [PMID: 36230547 PMCID: PMC9563402 DOI: 10.3390/cancers14194625] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Oral squamous cell carcinoma accounts for 95% of human head and neck squamous cell carcinoma cases. It is highly malignant and aggressive, with a poor prognosis and a 5-year survival rate of <50%. In recent years, basic and clinical studies have been performed on the role of the mitogen-activated protein kinase (MAPK) signaling pathway in oral cancer. The MAPK signaling pathway is activated in over 50% of human oral cancer cases. Herein, we review research progress on the MAPK signaling pathway and its potential therapeutic mechanisms and discuss its molecular targeting to explore its potential as a therapeutic strategy for oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Yuxi Cheng
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Juan Chen
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Yuxin Shi
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Xiaodan Fang
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
- Correspondence: (X.F.); (Z.T.)
| | - Zhangui Tang
- Xiangya Stomatological Hospital, Central South University, Changsha 410008, China
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
- Correspondence: (X.F.); (Z.T.)
| |
Collapse
|
12
|
Zhang Y, Wang Z, Wang R, Xia L, Cai Y, Tong F, Gao Y, Ding J, Wang X. Conditional knockout of ASK1 in microglia/macrophages attenuates epileptic seizures and long-term neurobehavioural comorbidities by modulating the inflammatory responses of microglia/macrophages. J Neuroinflammation 2022; 19:202. [PMID: 35941644 PMCID: PMC9361603 DOI: 10.1186/s12974-022-02560-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Apoptosis signal-regulating kinase 1 (ASK1) not only causes neuronal programmed cell death via the mitochondrial pathway but also is an essential component of the signalling cascade during microglial activation. We hypothesize that ASK1 selective deletion modulates inflammatory responses in microglia/macrophages(Mi/Mϕ) and attenuates seizure severity and long-term cognitive impairments in an epileptic mouse model. Methods Mi/Mϕ-specific ASK1 conditional knockout (ASK1 cKO) mice were obtained for experiments by mating ASK1flox/flox mice with CX3CR1creER mice with tamoxifen induction. Epileptic seizures were induced by intrahippocampal injection of kainic acid (KA). ASK1 expression and distribution were detected by western blotting and immunofluorescence staining. Seizures were monitored for 24 h per day with video recordings. Cognition, social and stress related activities were assessed with the Y maze test and the three-chamber social novelty preference test. The heterogeneous Mi/Mϕ status and inflammatory profiles were assessed with immunofluorescence staining and real-time polymerase chain reaction (q-PCR). Immunofluorescence staining was used to detect the proportion of Mi/Mϕ in contact with apoptotic neurons, as well as neuronal damage. Results ASK1 was highly expressed in Mi/Mϕ during the acute phase of epilepsy. Conditional knockout of ASK1 in Mi/Mϕ markedly reduced the frequency of seizures in the acute phase and the frequency of spontaneous recurrent seizures (SRSs) in the chronic phase. In addition, ASK1 conditional knockout mice displayed long-term neurobehavioral improvements during the Y maze test and the three-chamber social novelty preference test. ASK1 selective knockout mitigated neuroinflammation, as evidenced by lower levels of Iba1+/CD16+ proinflammatory Mi/Mϕ. Conditional knockout of ASK1 increased Mi/Mϕ proportion in contact with apoptotic neurons. Neuronal loss was partially restored by ASK1 selective knockout. Conclusion Conditional knockout of ASK1 in Mi/Mϕ reduced seizure severity, neurobehavioral impairments, and histological damage, at least via inhibiting proinflammatory microglia/macrophages responses. ASK1 in microglia/macrophages is a potential therapeutic target for inflammatory responses in epilepsy. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02560-5.
Collapse
Affiliation(s)
- Yiying Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhangyang Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Rongrong Wang
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lu Xia
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yiying Cai
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Fangchao Tong
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yanqin Gao
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Singh V, Huang E, Pathak V, Willard BB, Allende DS, Nagy LE. Phosphoproteomics identifies pathways underlying the role of receptor-interaction protein kinase 3 in alcohol-associated liver disease and uncovers apoptosis signal-regulating kinase 1 as a target. Hepatol Commun 2022; 6:2022-2041. [PMID: 35438255 PMCID: PMC9315126 DOI: 10.1002/hep4.1956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 01/21/2023] Open
Abstract
Receptor-interaction protein kinase 3 (RIP3), a critical determinant of the necroptotic pathway of programmed cell death, contributes to injury in murine models of alcohol-associated liver disease (ALD); however, the underlying mechanisms are unknown. We investigated the effect of chronic ethanol feeding on the hepatic phosphoproteome in C57BL/6 and RIP3-deficient (Rip3-/- ) mice, focusing on death receptor (DR) signaling pathways. C57BL/6 and Rip3-/- mice were fed an ethanol-containing liquid diet or pair-fed control diet. A label-free mass spectrometry-based approach identified differentially phosphorylated proteins that were mapped to pathways affected by ethanol and Rip3 genotype. Identified targets were validated in both the murine model of ALD and in liver tissue from patients with alcohol-associated hepatitis (AH) and healthy controls. Chronic ethanol dysregulated hepatic tumor necrosis factor-induced DR signaling pathways. Of particular importance, chronic ethanol feeding to C57BL/6 mice decreased the phosphorylation of apoptosis signal-regulating kinase 1 (ASK1) at serine (S)1036/S1040 (S1029/S1033 human), sites linked with the inhibition of ASK1 death-promoting activity. This decrease in phosphorylation of inhibitory sites was muted in Rip3-/- mice. Decreased phosphorylation at S1033 was also lower in liver of patients with severe AH compared to healthy controls, and phosphorylation at the ASK1 activation site (threonine [Thr]-838) was increased in patients with AH. The net impact of these changes in phosphorylation of ASK1 was associated with increased phosphorylation of p38, a downstream target of ASK1, in patients with AH and C57BL/6 but not Rip3-/- mice. Similarly, chronic ethanol feeding affected the c-Jun N-terminal kinase pathway in C57BL/6 but not Rip3-/- mice. Taken together, our data indicate that changes in inhibitory phosphorylation of ASK1 are an important target in ALD and suggest the involvement of noncanonical functions of Rip3 in ALD.
Collapse
Affiliation(s)
- Vaibhav Singh
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Emily Huang
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Vai Pathak
- Department of Quantitative Health SciencesCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Belinda B Willard
- Proteomics and Metabolomics CoreCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | | | - Laura E Nagy
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| |
Collapse
|
14
|
Crystallographic mining of ASK1 regulators to unravel the intricate PPI interfaces for the discovery of small molecule. Comput Struct Biotechnol J 2022; 20:3734-3754. [PMID: 35891784 PMCID: PMC9294202 DOI: 10.1016/j.csbj.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Protein seldom performs biological activities in isolation. Understanding the protein–protein interactions’ physical rewiring in response to pathological conditions or pathogen infection can help advance our comprehension of disease etiology, progression, and pathogenesis, which allow us to explore the alternate route to control the regulation of key target interactions, timely and effectively. Nonalcoholic steatohepatitis (NASH) is now a global public health problem exacerbated due to the lack of appropriate treatments. The most advanced anti-NASH lead compound (selonsertib) is withdrawn, though it is able to inhibit its target Apoptosis signal-regulating kinase 1 (ASK1) completely, indicating the necessity to explore alternate routes rather than complete inhibition. Understanding the interaction fingerprints of endogenous regulators at the molecular level that underpin disease formation and progression may spur the rationale of designing therapeutic strategies. Based on our analysis and thorough literature survey of the various key regulators and PTMs, the current review emphasizes PPI-based drug discovery’s relevance for NASH conditions. The lack of structural detail (interface sites) of ASK1 and its regulators makes it challenging to characterize the PPI interfaces. This review summarizes key regulators interaction fingerprinting of ASK1, which can be explored further to restore the homeostasis from its hyperactive states for therapeutics intervention against NASH.
Collapse
Key Words
- ASK1
- ASK1, Apoptosis signal-regulating kinase 1
- CFLAR, CASP8 and FADD-like apoptosis regulator
- CREG, Cellular repressor of E1A-stimulated genes
- DKK3, Dickkopf-related protein 3
- Interaction fingerprint
- NAFLD, Non-alcoholic fatty liver disease
- NASH
- NASH, Nonalcoholic steatohepatitis
- PPI, Protein-protein interaction
- PTM, Post-trancriptional modification
- PTMs
- Protein-protein interaction
- TNFAIP3, TNF Alpha Induced Protein 3
- TRAF2/6, Tumor necrosis factor receptor (TNFR)-associated factor2/6
- TRIM48, Tripartite Motif Containing 48
- TRX, Thioredoxin
- USP9X, Ubiquitin Specific Peptidase 9 X-Linked
Collapse
|
15
|
Jin Y, Tan Y, Zhao P, Guo Y, Chen S, Wu J, Ren Z. Glutathione S-transferase Mu 2 inhibits hepatic steatosis via ASK1 suppression. Commun Biol 2022; 5:326. [PMID: 35388144 PMCID: PMC8986781 DOI: 10.1038/s42003-022-03251-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 03/11/2022] [Indexed: 12/31/2022] Open
Abstract
Hepatic steatosis is the main characteristic of some liver metabolism diseases. However, unclear molecular mechanism of hepatic steatosis impedes the therapy of this hepatic steatosis. Glutathione-S-transferase mu 2 (GSTM2), as a member of phase II drug metabolizing enzymes (DMEs), regulates cellular antioxidant and detoxificant. GSTM2 was highly up-regulated in hepatic steatosis tissues and high-fat diet (HFD) fed mice. Loss-of-function GSTM2 mouse model demonstrated that GSTM2 protected mice from excess fat accumulation. Mechanistically, GSTM2 interacted with ASK1 and suppressed its phosphorylation and the activation of subsequent downstream p38-JNK signalling. Moreover, GSTM2 overexpression in the liver effectively ameliorated hepatic lipid accumulation. Therefore, we identified GSTM2 as an important negative regulator in progression of hepatic steatosis via both its detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling. This study showed potential therapeutic function of the DME in progression of hepatic steatosis. Jin et al. investigate the underlying mechanisms of hepatic steatosis and show that Glutathione-S-transferase mu 2 (GSTM2), which is a drug metabolizing enzyme (DME), is upregulated in hepatic steatosis tissue from mice fed with a high fat diet. They show that GSTM2 is a negative regulator of hepatic steatosis via both detoxification/antioxidant and inhibition of ASK1-p38/JNK signalling, which sheds light on its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China
| | - Yanjie Tan
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Pengxiang Zhao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Yu Guo
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Shilin Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China.
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China. .,Hubei Hongshan Laboratory, Wuhan, Hubei, 430070, PR China.
| |
Collapse
|
16
|
Zhang X, Yang H, Zeng S, Tian S, Hu S, Yang L, Ma T, Liu Z, Wan J, Zhong Y, Li H. Melanoma differentiation-Associated gene 5 protects against NASH in mice. Hepatology 2022; 75:924-938. [PMID: 34482560 DOI: 10.1002/hep.32139] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS NASH is a complicated disease characterized by hepatocyte steatosis, inflammation infiltration, and liver fibrosis. Accumulating evidence suggests that the innate immunity plays a key role in NASH progression. Here, we aimed to reveal the role of melanoma differentiation-associated gene 5 (MDA5, also known as Ifih1), a conventional innate immune regulator following viral infection, in the progression of NASH and investigate its underlying mechanism. APPROACH AND RESULTS We first examined the expression of MDA5 and found that MDA5 was markedly down-regulated in the livers with NASH in human individuals and mice models. MDA5 overexpression significantly inhibits the free fatty acid-induced lipid accumulation and inflammation in hepatocyte in vitro, whereas MDA5 knockdown promotes hepatocyte lipotoxicity. Using hepatocyte-specific Mda5 gene knockout and transgenic mice, we found that diet-induced hepatic steatosis, inflammation, and liver fibrosis were markedly exacerbated by Mda5 deficiency but suppressed by Mda5 overexpression. Mechanistically, we found that the activation of apoptosis signal-regulating kinase 1 (ASK1)-mitogen-activated protein kinase pathway was significantly inhibited by MDA5 but enhanced by MDA5 deletion. We further validated that MDA5 directly interacted with ASK1 and suppressed its N-terminal dimerization. Importantly, blockage of ASK1 with adenovirus-expressing dominant negative ASK1 obviously reversed the lipid accumulation and ASK1 pathway activation when Mda5 was knocked out. CONCLUSIONS These data indicate that MDA5 is an essential suppressor in NASH. The findings support MDA5 as a regulator of ASK1 and a promising therapeutic target for NASH.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of EducationGannan Medical UniversityGanzhouChina.,Gannan Innovation and Translational Medicine Research InstituteGanzhouChina
| | - Hailong Yang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Shan Zeng
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of EducationGannan Medical UniversityGanzhouChina.,Gannan Innovation and Translational Medicine Research InstituteGanzhouChina
| | - Song Tian
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Sha Hu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Ling Yang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Tengfei Ma
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Zhen Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Juan Wan
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina
| | - Yiming Zhong
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of EducationGannan Medical UniversityGanzhouChina.,Gannan Innovation and Translational Medicine Research InstituteGanzhouChina
| | - Hongliang Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina.,Institute of Model AnimalWuhan UniversityWuhanChina.,Medical Science Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
17
|
Obsilova V, Honzejkova K, Obsil T. Structural Insights Support Targeting ASK1 Kinase for Therapeutic Interventions. Int J Mol Sci 2021; 22:ijms222413395. [PMID: 34948191 PMCID: PMC8705584 DOI: 10.3390/ijms222413395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/22/2022] Open
Abstract
Apoptosis signal-regulating kinase (ASK) 1, a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family, modulates diverse responses to oxidative and endoplasmic reticulum (ER) stress and calcium influx. As a crucial cellular stress sensor, ASK1 activates c-Jun N-terminal kinases (JNKs) and p38 MAPKs. Their excessive and sustained activation leads to cell death, inflammation and fibrosis in various tissues and is implicated in the development of many neurological disorders, such as Alzheimer’s, Parkinson’s and Huntington disease and amyotrophic lateral sclerosis, in addition to cardiovascular diseases, diabetes and cancer. However, currently available inhibitors of JNK and p38 kinases either lack efficacy or have undesirable side effects. Therefore, targeted inhibition of their upstream activator, ASK1, stands out as a promising therapeutic strategy for treating such severe pathological conditions. This review summarizes recent structural findings on ASK1 regulation and its role in various diseases, highlighting prospects for ASK1 inhibition in the treatment of these pathologies.
Collapse
Affiliation(s)
- Veronika Obsilova
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
- Correspondence: (V.O.); (T.O.); Tel.: +420-325-87-3513 (V.O.); +420-22-195-1303 (T.O.)
| | - Karolina Honzejkova
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 12843 Prague, Czech Republic;
| | - Tomas Obsil
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 12843 Prague, Czech Republic;
- Correspondence: (V.O.); (T.O.); Tel.: +420-325-87-3513 (V.O.); +420-22-195-1303 (T.O.)
| |
Collapse
|
18
|
Sun A, Zhu J, Xia S, Li Y, Wu T, Shao G, Yang W, Lin Q. MEKK5 Interacts with and Negatively Regulates the E3 Ubiquitin Ligase NEDD4 for Mediating Lung Cancer Cell Migration. Life (Basel) 2021; 11:life11111153. [PMID: 34833029 PMCID: PMC8620495 DOI: 10.3390/life11111153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Our previous studies have shown that the HECT E3 ubiquitin ligase NEDD4 and kinase MEKK5 both play an essential role in lung cancer migration. A report predicts that MEKK5 may be ubiquitinated by NEDD4; however, interaction of MEKK5 with NEDD4 and ubiquitination of MEKK5 by NEDD4 have not been characterized. In this report, we show that NEDD4 interacts with MEKK5 through a conserved WW3 domain by the co-immunoprecipitation and the GST-pulldown assays. The ubiquitination assay indicates that MEKK5 is not a ubiquitination substrate of NEDD4, but negatively regulates NEDD4-mediated ubiquitination. Furthermore, overexpression of MEKK5 significantly reduced the NEDD4-promoted lung cancer cell migration. Taken together, our studies have defined an inhibitory role of MEKK5 in regulation of NEDD4-mediated ubiquitination.
Collapse
|
19
|
Zhang BB, Gao L, Yang Q, Liu Y, Yu XY, Shen JH, Zhang WC, Han ZY, Chen SZ, Guo S. Role of GALNT4 in protecting against cardiac hypertrophy through ASK1 signaling pathway. Cell Death Dis 2021; 12:980. [PMID: 34675184 PMCID: PMC8531281 DOI: 10.1038/s41419-021-04222-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Pathological myocardial hypertrophy is regulated by multiple pathways. However, its underlying pathogenesis has not been fully explored. The goal of this work was to elucidate the function of polypeptide N-acetylgalactosaminyltransferase 4 (GALNT4) in myocardial hypertrophy and its underlying mechanism of action. We illustrated that GALNT4 was upregulated in the models of hypertrophy. Two cardiac hypertrophy models were established through partial transection of the aorta in GALNT4-knockout (GALNT4-KO) mice and adeno-associated virus 9-GALNT4 (AAV9-GALNT4) mice. The GALNT4-KO mice demonstrated accelerated cardiac hypertrophy, dysfunction, and fibrosis, whereas the opposite phenotype was observed in AAV9-GALNT4 mice. Similarly, GALNT4 overexpression mitigated the degree of phenylephrine-induced cardiomyocyte hypertrophy in vitro whereas GALNT4 knockdown aggravated the hypertrophy. In terms of mechanism, GALNT4 deficiency increased the phosphorylation and activation of ASK1 and its downstream targets (JNK and p38), whereas GALNT4 overexpression inhibited activation of the ASK1 pathway. Furthermore, we demonstrated that GALNT4 can directly bind to ASK1 inhibiting its N-terminally mediated dimerization and the subsequent phosphorylation of ASK1. Finally, an ASK1 inhibitor (iASK1) was able to reverse the effects of GALNT4 in vitro. In summary, GALNT4 may serve as a new regulatory factor and therapeutic target by blocking the activation of the ASK1 signaling cascade.
Collapse
Affiliation(s)
- 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
| | - Qin Yang
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Yuan Liu
- 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
| | - Wen-Cai Zhang
- 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.
| | - Shao-Ze Chen
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China.
- Huanggang Institute of Translational Medicine, Huanggang, China.
| | - Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, China.
| |
Collapse
|
20
|
Li X, Ding Z, Wu Z, Xu Y, Yao H, Lin K. Targeting the TGF-β signaling pathway for fibrosis therapy: a patent review (2015-2020). Expert Opin Ther Pat 2021; 31:723-743. [PMID: 33645365 DOI: 10.1080/13543776.2021.1896705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Fibrosis is a serious disease that occurs in many organs, such as kidney, liver and lung. The deterioration of these organs ultimately leads to death. Due to the complex mechanisms of fibrosis, research and development of antifibrotic drugs is difficult. One solution is to focus on core pathways, one of which is the TGF-β signaling pathway. In virtually every type of fibrosis, TGF-β signaling is recognized as a critical pathway. AREA COVERED This review discusses patents on active molecules related to the TGF-β signaling. Molecules targeting components related to the activation of TGF-β are introduced. Several strategies preventing signal propagation from active TGF-β to downstream targets are also introduced, including TGF-β antibodies, TGF-β ligand traps, and inhibitors of TGF-β receptor kinases. Finally, molecules affecting downstream targets in both canonical and noncanonical TGF-β signaling pathways are described. EXPERT OPINION Since the approval of pirfenidone, targeting TGF-β signaling has been anticipated as an effective therapy for fibrosis. The potential of this therapy has been further supported by emerging patents on the TGF-β signaling. This pathway can be entirely inhibited, from the activation of TGF-β to downstream signaling. Inhibiting TGF-β signaling is expected to provide more effective treatments for fibrosis.
Collapse
Affiliation(s)
- Xuanyi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ziang Ding
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zixuan Wu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yinqiu Xu
- Department of Pharmacy, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Hequan Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kejiang Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
21
|
Suzuki Y, Asami M, Takahashi D, Sakane F. Diacylglycerol kinase η colocalizes and interacts with apoptosis signal-regulating kinase 3 in response to osmotic shock. Biochem Biophys Rep 2021; 26:101006. [PMID: 33997319 PMCID: PMC8100535 DOI: 10.1016/j.bbrep.2021.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 11/29/2022] Open
Abstract
Diacylglycerol kinase (DGK) η translocates from the cytoplasm to punctate vehicles via osmotic shock. Apoptosis signal-regulating kinase (ASK) 3 (MAP kinase kinase kinase (MAPKKK) 15) is also reported to respond to osmotic shock. Therefore, in the present study, we examined the subcellular localization of DGKη and ASK3 expressed in COS-7 cells under osmotic stress. We found that DGKη was almost completely colocalized with ASK3 in punctate structures in response to osmotic shock. In contrast, DGKδ, which is closely related to DGKη structurally, was not colocalized with ASK3, and DGKη failed to colocalize with another MAPKKK, C-Raf, even under osmotic stress. The structures in which DGKη and ASK3 localized were not stained with stress granule makers. Notably, DGKη strongly interacted with ASK3 in an osmotic shock-dependent manner. These results indicate that DGKη and ASK3 undergo osmotic shock-dependent colocalization and associate with each other in specialized structures. DGKη translocates from the cytoplasm to punctate vehicles via osmotic stress. DGKη colocalizes with ASK3 in punctate vehicles in response to osmotic shock. DGKη interacts with ASK3 in response to osmotic shock. The punctate vesicles are unique and specialized for DGKη and ASK3.
Collapse
Affiliation(s)
- Yuji Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Maho Asami
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Daisuke Takahashi
- Department of Pharmaceutical Health Care and Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| |
Collapse
|
22
|
Esteban-Collado J, Corominas M, Serras F. Nutrition and PI3K/Akt signaling are required for p38-dependent regeneration. Development 2021; 148:258580. [PMID: 33913483 DOI: 10.1242/dev.197087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/29/2021] [Indexed: 12/22/2022]
Abstract
Regeneration after damage requires early signals to trigger the tissue repair machinery. Reactive oxygen species (ROS) act as early signals that are sensed by the MAP3 kinase Ask1, which in turn activates by phosphorylation the MAP kinases p38 and JNK. The sustained or high activation of these kinases can result in apoptosis, whereas short or low activation can promote regeneration. Using the Ask1-dependent regeneration program, we demonstrate in Drosophila wing that PI3K/Akt signaling is necessary for Ask1 to activate p38, but not JNK. In addition, nutrient restriction or mutations that target Ser83 of the Drosophila Ask1 protein, a PI3K/Akt-sensitive residue, block regeneration. However, these effects can be reversed by the ectopic activation of p38, but not of JNK. Our results demonstrate that Ask1 controls the activation of p38 through Ser83, and that the phosphorylation of p38 during regeneration is nutrient sensitive. This mechanism is important for discriminating between p38 and JNK in the cells involved in tissue repair and regenerative growth.
Collapse
Affiliation(s)
- José Esteban-Collado
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), Diagonal 643, 08028 Barcelona, Spain
| |
Collapse
|
23
|
Xiang M, Yang F, Zhou Y, Li W, Zou Y, Ye P, Zhu L, Wang PX, Chen M. LITAF acts as a novel regulator for pathological cardiac hypertrophy. J Mol Cell Cardiol 2021; 156:82-94. [PMID: 33823186 DOI: 10.1016/j.yjmcc.2021.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/04/2021] [Accepted: 03/25/2021] [Indexed: 11/19/2022]
Abstract
Pathological hypertrophy generally progresses to heart failure. Exploring effective and promising therapeutic targets might lead to progress in preventing its detrimental outcomes. Our current knowledge about lipopolysaccharide-induced tumor necrosis factor-α factor (LITAF) is mainly limited to regulate inflammation. However, the role of LITAF in other settings that are not that relevant to inflammation, such as cardiac remodeling and heart failure, remains largely unknown. In the present study, we found that the expression of LITAF decreased in hypertrophic hearts and cardiomyocytes. Meanwhile, LITAF protected cultured neonatal rat cardiomyocytes against phenylephrine-induced hypertrophy. Moreover, using LITAF knockout mice, we demonstrated that LITAF deficiency exacerbated cardiac hypertrophy and fibrosis compared with wild-type mice. Mechanistically, LITAF directly binds to the N-terminal of ASK1, thus disrupting the dimerization of ASK1 and blocking ASK1 activation, ultimately inhibiting ASK1-JNK/p38 signaling over-activation and protecting against cardiac hypertrophy. Furthermore, AAV9-mediated LITAF overexpression attenuated cardiac hypertrophy in vivo. Conclusions: Our findings uncover the novel role of LITAF as a negative regulator of cardiac remodeling. Targeting the interaction between LITAF and ASK1 could be a promising therapeutic strategy for pathological cardiac remodeling.
Collapse
Affiliation(s)
- Mei Xiang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Feiyan Yang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yi Zhou
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Weijuan Li
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yuanlin Zou
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ling Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Pi-Xiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China.
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China.
| |
Collapse
|
24
|
Shi Y, Bai J, Dang Y, Bai Q, Zheng R, Chen J, Li Z. Protection of apigenin against acrylonitrile-induced sperm and testis injury in rats: involvement of activation of ASK1-JNK/p38 signaling pathway. Toxicol Res (Camb) 2021; 10:159-168. [PMID: 33884167 DOI: 10.1093/toxres/tfab017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/14/2021] [Accepted: 02/07/2021] [Indexed: 11/14/2022] Open
Abstract
This study aims to clarify if apigenin (AP) could play a pivotal role in attenuating acrylonitrile (ACN)-induced sperm and testis injury by inhibiting ASK1-JNK/p38 signaling pathway. Male Sprague-Dawley rats were randomly divided into five groups: a control group (corn oil), an ACN group (ACN 46 mg kg-1), an ACN + AP1 group (ACN + AP 117 mg kg-1), an ACN + AP2 group (ACN + AP 234 mg kg-1) and an ACN + AP3 group (ACN + AP 351 mg kg-1). The ACN + AP groups were given ACN by gavage after a pretreatment with different dosages of AP for 30 min, whereas the rats in the control group received an equivalent volume of corn oil. The gavage was conducted for 6 days per week in 4 weeks. The results showed that AP reduced sperm deformity rate and DNA fragment index and attenuated the testicular injury induced by ACN. AP could also alleviate oxidative stress, downregulate ASK1-JNK/p38 signaling pathway and eventually inhibit mitochondria-mediated testicular apoptosis. In brief, AP could dampen oxidative stress thereby inhibiting testicular apoptosis mediated by ASK1-JNK/p38 signaling pathway, alleviating ACN-induced sperm and testis injury and exerting a protective effect on male reproductive system.
Collapse
Affiliation(s)
- Ying Shi
- Lanzhou Maternal and Child Health Care Hospital, Lanzhou 730030, China
| | - Jin Bai
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Yuhui Dang
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qingli Bai
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Rong Zheng
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jia Chen
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Zhilan Li
- Institute of Maternal, Child and Adolescent Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
25
|
Vellino S, Oddou C, Rivier P, Boyault C, Hiriart-Bryant E, Kraut A, Martin R, Coute Y, Knölker HJ, Valverde MA, Albigès-Rizo C, Destaing O. Cross-talk between the calcium channel TRPV4 and reactive oxygen species interlocks adhesive and degradative functions of invadosomes. J Cell Biol 2021; 220:211651. [PMID: 33399853 PMCID: PMC7788461 DOI: 10.1083/jcb.201910079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 07/23/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Invadosomes support cell invasion by coupling both acto-adhesive and extracellular matrix degradative functions, which are apparently antagonistic. β1-integrin dynamics regulate this coupling, but the actual sensing mechanism and effectors involved have not yet been elucidated. Using genetic and reverse genetic approaches combined with biochemical and imaging techniques, we now show that the calcium channel TRPV4 colocalizes with β1-integrins at the invadosome periphery and regulates its activation and the coupling of acto-adhesive and degradative functions. TRPV4-mediated regulation of podosome function depends on its ability to sense reactive oxygen species (ROS) in invadosomes' microenvironment and involves activation of the ROS/calcium-sensitive kinase Ask1 and binding of the motor MYO1C. Furthermore, disease-associated TRPV4 gain-of-function mutations that modulate ECM degradation are also implicated in the ROS response, which provides new perspectives in our understanding of the pathophysiology of TRPV4 channelopathies.
Collapse
Affiliation(s)
- Sanela Vellino
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Christiane Oddou
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Paul Rivier
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Cyril Boyault
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Edwige Hiriart-Bryant
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Alexandra Kraut
- Laboratoire EDyP, Institute of Biosciences and Biotechnologies of Grenoble-Biologie à Grande Echelle, Commissariat à l'Énergie Atomique Grenoble, Grenoble, France
| | - René Martin
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Yohann Coute
- Laboratoire EDyP, Institute of Biosciences and Biotechnologies of Grenoble-Biologie à Grande Echelle, Commissariat à l'Énergie Atomique Grenoble, Grenoble, France
| | | | - Miguel A. Valverde
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Corinne Albigès-Rizo
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France
| | - Olivier Destaing
- Dynamique des systèmes d'adhérence, Institut for Advanced Biosciences, Centre de Recherche University Grenoble Alpes/INSERM U1209/Centre National de la Recherche Scientifique Unité mixte de recherche 5309, La Tronche, France,Correspondence to Olivier Destaing:
| |
Collapse
|
26
|
Yarmohammadi F, Rezaee R, Haye AW, Karimi G. Endoplasmic reticulum stress in doxorubicin-induced cardiotoxicity may be therapeutically targeted by natural and chemical compounds: A review. Pharmacol Res 2020; 164:105383. [PMID: 33348022 DOI: 10.1016/j.phrs.2020.105383] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent with marked, dose-dependent cardiotoxicity that leads to tachycardia, atrial and ventricular arrhythmia, and irreversible heart failure. Induction of the endoplasmic reticulum (ER) which plays a major role in protein folding and calcium homeostasis was reported as a key contributor to cardiac complications of DOX. This article reviews several chemical compounds that have been shown to regulate DOX-induced inflammation, apoptosis, and autophagy via inhibition of ER stress signaling pathways, such as the IRE1α/ASK1/JNK, IRE1α/JNK/Beclin-1, and CHOP pathways.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Haye
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Identification of Uncharacterized Components of Prokaryotic Immune Systems and Their Diverse Eukaryotic Reformulations. J Bacteriol 2020; 202:JB.00365-20. [PMID: 32868406 DOI: 10.1128/jb.00365-20] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022] Open
Abstract
Nucleotide-activated effector deployment, prototyped by interferon-dependent immunity, is a common mechanistic theme shared by immune systems of several animals and prokaryotes. Prokaryotic versions include CRISPR-Cas with the CRISPR polymerase domain, their minimal variants, and systems with second messenger oligonucleotide or dinucleotide synthetase (SMODS). Cyclic or linear oligonucleotide signals in these systems help set a threshold for the activation of potentially deleterious downstream effectors in response to invader detection. We establish such a regulatory mechanism to be a more general principle of immune systems, which can also operate independently of such messengers. Using sensitive sequence analysis and comparative genomics, we identify 12 new prokaryotic immune systems, which we unify by this principle of threshold-dependent effector activation. These display regulatory mechanisms paralleling physiological signaling based on 3'-5' cyclic mononucleotides, NAD+-derived messengers, two- and one-component signaling that includes histidine kinase-based signaling, and proteolytic activation. Furthermore, these systems allowed the identification of multiple new sensory signal sensory components, such as a tetratricopeptide repeat (TPR) scaffold predicted to recognize NAD+-derived signals, unreported versions of the STING domain, prokaryotic YEATS domains, and a predicted nucleotide sensor related to receiver domains. We also identify previously unrecognized invader detection components and effector components, such as prokaryotic versions of the Wnt domain. Finally, we show that there have been multiple acquisitions of unidentified STING domains in eukaryotes, while the TPR scaffold was incorporated into the animal immunity/apoptosis signal-regulating kinase (ASK) signalosome.IMPORTANCE Both prokaryotic and eukaryotic immune systems face the dangers of premature activation of effectors and degradation of self-molecules in the absence of an invader. To mitigate this, they have evolved threshold-setting regulatory mechanisms for the triggering of effectors only upon the detection of a sufficiently strong invader signal. This work defines general templates for such regulation in effector-based immune systems. Using this, we identify several previously uncharacterized prokaryotic immune mechanisms that accomplish the regulation of downstream effector deployment by using nucleotide, NAD+-derived, two-component, and one-component signals paralleling physiological homeostasis. This study has also helped identify several previously unknown sensor and effector modules in these systems. Our findings also augment the growing evidence for the emergence of key animal immunity and chromatin regulatory components from prokaryotic progenitors.
Collapse
|
28
|
Mishra S, Dunkerly-Eyring BL, Keceli G, Ranek MJ. Phosphorylation Modifications Regulating Cardiac Protein Quality Control Mechanisms. Front Physiol 2020; 11:593585. [PMID: 33281625 PMCID: PMC7689282 DOI: 10.3389/fphys.2020.593585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many forms of cardiac disease, including heart failure, present with inadequate protein quality control (PQC). Pathological conditions often involve impaired removal of terminally misfolded proteins. This results in the formation of large protein aggregates, which further reduce cellular viability and cardiac function. Cardiomyocytes have an intricately collaborative PQC system to minimize cellular proteotoxicity. Increased expression of chaperones or enhanced clearance of misfolded proteins either by the proteasome or lysosome has been demonstrated to attenuate disease pathogenesis, whereas reduced PQC exacerbates pathogenesis. Recent studies have revealed that phosphorylation of key proteins has a potent regulatory role, both promoting and hindering the PQC machinery. This review highlights the recent advances in phosphorylations regulating PQC, the impact in cardiac pathology, and the therapeutic opportunities presented by harnessing these modifications.
Collapse
Affiliation(s)
- Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brittany L Dunkerly-Eyring
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
29
|
Hattori K, Wakatsuki H, Sakauchi C, Furutani S, Sugawara S, Hatta T, Natsume T, Ichijo H. β-adrenergic receptor signaling evokes the PKA-ASK axis in mature brown adipocytes. PLoS One 2020; 15:e0232645. [PMID: 33108364 PMCID: PMC7591029 DOI: 10.1371/journal.pone.0232645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/03/2020] [Indexed: 12/31/2022] Open
Abstract
Boosting energy expenditure by harnessing the activity of brown adipocytes is a promising strategy for combatting the global epidemic of obesity. Many studies have revealed that the β3-adrenergic receptor agonist is a potent activator of brown adipocytes, even in humans, and PKA and p38 MAPK have been demonstrated for regulating the transcription of a wide range of critical genes such as Ucp1. We previously revealed that the PKA-ASK1-p38 axis is activated in immature brown adipocytes and contributes to functional maturation. However, the downstream mechanisms of PKA that initiate the p38 MAPK cascade are still mostly unknown in mature brown adipocytes. Here, we identified the ASK family as a crucial signaling molecule bridging PKA and MAPK in mature brown adipocytes. Mechanistically, the phosphorylation of ASK1 at threonine 99 and serine 993 is critical in PKA-dependent ASK1 activation. Additionally, PKA also activates ASK2, which contributes to MAPK regulation. These lines of evidence provide new details for tailoring a βAR-dependent brown adipocyte activation strategy.
Collapse
Affiliation(s)
- Kazuki Hattori
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| | - Hiroaki Wakatsuki
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Chihiro Sakauchi
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shotaro Furutani
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Sho Sugawara
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Hatta
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Hidenori Ichijo
- The Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail: (KH); (HI)
| |
Collapse
|
30
|
Li F, Yang Y, Xue C, Tan M, Xu L, Gao J, Xu L, Zong J, Qian W. Zinc Finger Protein ZBTB20 protects against cardiac remodelling post-myocardial infarction via ROS-TNFα/ASK1/JNK pathway regulation. J Cell Mol Med 2020; 24:13383-13396. [PMID: 33063955 PMCID: PMC7701508 DOI: 10.1111/jcmm.15961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/03/2023] Open
Abstract
This study aims to determine the efficacy of Zinc finger protein ZBTB20 in treatment of post‐infarction cardiac remodelling. For this purpose, left anterior descending (LAD) ligation was operated on mice to induce myocardial infarction (MI) with sham control group as contrast and adeno‐associated virus (AAV9) system was used to deliver ZBTB20 to mouse heart by myocardial injection with vehicle‐injected control group as contrast two weeks before MI surgery. Then four weeks after MI, vehicle‐treated mice with left ventricular (LV) remodelling underwent deterioration of cardiac function, with symptoms of hypertrophy, interstitial fibrosis, inflammation and apoptosis. The vehicle‐injected mice also showed increase of infarct size and decrease of survival rate. Meanwhile, the ZBTB20‐overexpressed mice displayed improvement after MI. Moreover, the anti‐apoptosis effect of ZBTB20 was further confirmed in H9c2 cells subjected to hypoxia in vitro. Further study suggested that ZBTB20 exerts cardioprotection by inhibiting tumour necrosis factor α/apoptosis signal‐regulating kinase 1 (ASK1)/c‐Jun N‐terminal kinase 1/2 (JNK1/2) signalling, which was confirmed by shRNA‐JNK adenoviruses transfection or a JNK activator in vitro as well as ASK1 overexpression in vivo. In summary, our data suggest that ZBTB20 could alleviate cardiac remodelling post‐MI. Thus, administration of ZBTB20 can be considered as a promising treatment strategy for heart failure post‐MI. Significance Statement: ZBTB20 could alleviate cardiac remodelling post‐MI via inhibition of ASK1/JNK1/2 signalling.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Yiming Yang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Chuanyou Xue
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Mengtong Tan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Lu Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Jianbo Gao
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Luhong Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Jing Zong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Wenhao Qian
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
31
|
Peroxiredoxin-1 Overexpression Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibiting Oxidative Stress and Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2405135. [PMID: 32802259 PMCID: PMC7411498 DOI: 10.1155/2020/2405135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/19/2020] [Accepted: 07/04/2020] [Indexed: 02/06/2023]
Abstract
Background. Previous research has shown that peroxiredoxin 1 (Prdx1) is an important modulator of physiological and pathophysiological cardiovascular events. This study is aimed at investigating the role and underlying mechanism of Prdx1 in doxorubicin- (DOX-) induced cardiotoxicity. Cardiac-specific expression of Prdx1 was induced in mice, and the mice received a single dose of DOX (15 mg/kg) to generate cardiotoxicity. First, our study demonstrated that Prdx1 expression was upregulated in the heart and in cardiomyocytes after DOX treatment. Second, we provided direct evidence that Prdx1 overexpression ameliorated DOX-induced cardiotoxicity by attenuating oxidative stress and cardiomyocyte apoptosis. Mechanistically, we found that DOX treatment increased the phosphorylation level of apoptosis signal-regulating kinase-1 (ASK1) and the downstream protein p38 in the heart and in cardiomyocytes, and these effects were decreased by Prdx1 overexpression. In contrast, inhibiting Prdx1 promoted DOX-induced cardiac injury via the ASK1/p38 pathway. These results suggest that Prdx1 may be an effective therapeutic option to prevent DOX-induced cardiotoxicity.
Collapse
|
32
|
Calcium-induced dissociation of CIB1 from ASK1 regulates agonist-induced activation of the p38 MAPK pathway in platelets. Biochem J 2020; 476:2835-2850. [PMID: 31530712 DOI: 10.1042/bcj20190410] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase (MAPKKK) that regulates activation of the c-Jun N-terminal kinase (JNK)- and p38-stress response pathways leading to apoptosis in nucleated cells. We have previously shown that ASK1 is expressed in platelets and regulates agonist-induced platelet activation and thrombosis. However, the mechanism by which platelet agonists cause activation of ASK1 is unknown. Here, we show that in platelets agonist-induced activation of p38 is exclusively dependent on ASK1. Both thrombin and collagen were able to activate ASK1/p38. Activation of ASK1/p38 was strongly dependent on thromboxane A2 (TxA2) and ADP. Agonist-induced ASK1 activation is blocked by inhibition of phospholipase C (PLC) β/γ activity or by chelating intracellular Ca2+. Furthermore, treatment of platelets with thapsigargin or Ca2+ ionophore robustly induced ASK1/p38 activation. In addition, calcium and integrin-binding protein 1 (CIB1), a Ca2+-dependent negative regulator of ASK1, associates with ASK1 in resting platelets and is dissociated upon platelet activation by thrombin. Dissociation of CIB1 corresponds with ASK1 binding to tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6) and the autophosphorylation of ASK1 Thr838 within the catalytic domain results in full activation of ASK1. Furthermore, genetic ablation of Cib1 in mice augments agonist-induced Ask1/p38 activation. Together our results suggest that in resting platelets ASK1 is bound to CIB1 at low Ca2+ concentrations. Agonist-induced platelet activation causes an increase in intracellular Ca2+ concentration that leads to the dissociation of CIB1 from ASK1, allowing for proper dimerization through ASK1 N-terminal coiled-coil (NCC) domains.
Collapse
|
33
|
Xu X, Zhang Z, Lu Y, Sun Q, Liu Y, Liu Q, Tian W, Yin Y, Yu H, Sun B. ARRB1 ameliorates liver ischaemia/reperfusion injury via antagonizing TRAF6-mediated Lysine 6-linked polyubiquitination of ASK1 in hepatocytes. J Cell Mol Med 2020; 24:7814-7828. [PMID: 32445435 PMCID: PMC7348167 DOI: 10.1111/jcmm.15412] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatic ischaemia/reperfusion (I/R) injury is a major clinical problem during liver surgical procedures, which usually lead to early transplantation failure and higher organ rejection rate, and current effective therapeutic strategies are still limited. Therefore, in‐depth exploring of the molecular mechanisms underlying liver I/R injury is key to the development of new therapeutic methods. β‐arrestins are multifunctional proteins serving as important signalling scaffolds in numerous physiopathological processes, including liver‐specific diseases. However, the role and underlying mechanism of β‐arrestins in hepatic I/R injury remain largely unknown. Here, we showed that only ARRB1, but not ARRB2, was down‐regulated during liver I/R injury. Hepatocyte‐specific overexpression of ARRB1 significantly ameliorated liver damage, as demonstrated by decreases in serum aminotransferases, hepatocellular necrosis and apoptosis, infiltrating inflammatory cells and secretion of pro‐inflammatory cytokines relative to control mice, whereas experiments with ARRB1 knockout mice gotten opposite effects. Mechanistically, ARRB1 directly interacts with ASK1 in hepatocytes and inhibits its TRAF6‐mediated Lysine 6‐linked polyubiquitination, which then prevents the activation of ASK1 and its downstream signalling pathway during hepatic I/R injury. In addition, inhibition of ASK1 remarkably abolished the disruptive effect result from ARRB1 deficiency in liver I/R injury in vivo, indicating that ASK1 was required for ARRB1 function in hepatic I/R injury. In conclusion, we proposed that ARRB1 is a novel protective regulator during liver I/R injury, and modulation of the regulatory axis between ARRB1 and ASK1 could be a novel therapeutic strategy to prevent this pathological process.
Collapse
Affiliation(s)
- Xiaoliang Xu
- School of Medicine, Southeast University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zechuan Zhang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yijun Lu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qikai Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yang Liu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiaoyu Liu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenfang Tian
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Yin
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hailong Yu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Beicheng Sun
- School of Medicine, Southeast University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
34
|
Kiroski I, Jiang Y, Gavrilovici C, Gao F, Lee S, Scantlebury MH, Vandal M, Park SK, Tsai LH, Teskey GC, Rho JM, Nguyen MD. Reelin Improves Cognition and Extends the Lifespan of Mutant Ndel1 Mice with Postnatal CA1 Hippocampus Deterioration. Cereb Cortex 2020; 30:4964-4978. [PMID: 32328622 DOI: 10.1093/cercor/bhaa088] [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: 12/11/2019] [Revised: 02/25/2020] [Accepted: 03/21/2020] [Indexed: 01/01/2023] Open
Abstract
The glycoprotein Reelin maintains neuronal positioning and regulates neuronal plasticity in the adult brain. Reelin deficiency has been associated with neurological diseases. We recently showed that Reelin is depleted in mice with a targeted disruption of the Ndel1 gene in forebrain postnatal excitatory neurons (Ndel1 conditional knockout (CKO)). Ndel1 CKO mice exhibit fragmented microtubules in CA1 pyramidal neurons, profound deterioration of the CA1 hippocampus and a shortened lifespan (~10 weeks). Here we report that Ndel1 CKO mice (of both sexes) experience spatial learning and memory deficits that are associated with deregulation of neuronal cell adhesion, plasticity and neurotransmission genes, as assessed by genome-wide transcriptome analysis of the hippocampus. Importantly, a single injection of Reelin protein in the hippocampus of Ndel1 CKO mice improves spatial learning and memory function and this is correlated with reduced intrinsic hyperexcitability of CA1 pyramidal neurons, and normalized gene deregulation in the hippocampus. Strikingly, when treated with Reelin, Ndel1 CKO animals that die from an epileptic phenotype, live twice as long as nontreated, or vehicle-treated CKO animals. Thus, Reelin confers striking beneficial effects in the CA1 hippocampus, and at both behavioral and organismal levels.
Collapse
Affiliation(s)
- Ivana Kiroski
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Yulan Jiang
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Cezar Gavrilovici
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children's Hospital San Diego, 3020 Children's Way, MC 5009, San Diego, California 92123, USA
| | - Fan Gao
- The Picower Institute for Learning and Memory, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, Boston, USA
| | - Sukyoung Lee
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Morris H Scantlebury
- Departments of Pediatrics and Clinical Neurosciences, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Milene Vandal
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Li-Huei Tsai
- The Picower Institute for Learning and Memory, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, Boston, USA
| | - G Campbell Teskey
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Jong M Rho
- Departments of Neurosciences & Pediatrics, University of California San Diego, Rady Children's Hospital San Diego, 3020 Children's Way, MC 5009, San Diego, California 92123, USA
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology & Anatomy, and Biochemistry & Molecular Biology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| |
Collapse
|
35
|
Hirata Y. [Reactive Oxygen Species (ROS) Signaling: Regulatory Mechanisms and Pathophysiological Roles]. YAKUGAKU ZASSHI 2020; 139:1235-1241. [PMID: 31582606 DOI: 10.1248/yakushi.19-00141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules generated during mitochondrial respiration and under various environmental stresses, and cause damage to DNA, proteins, and lipids, which is linked to a wide variety of pathologies. However, recent studies have revealed the physiological importance of ROS as signaling molecules, which play crucial roles in the maintenance of cellular functions and homeostasis. According to the extent and duration of ROS generation, ROS-mediated oxidation-reduction (redox) signaling (ROS signaling) is tightly regulated by various molecules and post-translational modifications (PTMs), for inducing appropriate cellular responses. Dysregulation of ROS signaling causes cellular malfunctions, which are also linked to various diseases, such as cancer, neurodegeneration and inflammatory diseases. In this review, we focus on a ROS-responsive protein kinase apoptosis signal-regulating kinase 1 (ASK1) that belongs to the mitogen-activated protein (MAP) kinase kinase kinase (MAP3K) family, and activates the c-jun N-terminal kinase (JNK) and p38 MAP kinase pathways, which consequently induces various cellular responses, including apoptosis and inflammation. Here, we introduce a novel regulatory mechanism and the pathophysiological significance of ASK1 activation. We found that an E3 ubiquitin ligase TRIM48 orchestrates fine-tuning of ROS-induced ASK1 activation mediated by multiple types of PTMs, including ubiquitination, methylation, and phosphorylation. We also found that trans-fatty acids (TFAs) enhance ROS-dependent ASK1 activation induced by extracellular ATP, a damage-associated molecular pattern (DAMP), and thereby promotes apoptosis, which possibly contributes to the pathogenesis of TFA-related diseases including atherosclerosis. Thus, this review provides recent advances in the study of ROS signaling, especially ROS-ASK1 signaling pathway.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
36
|
Morales-Prieto N, López de Lerma N, Pacheco IL, Huertas-Abril PV, Pérez J, Peinado R, Abril N. Protective effect of Pedro-Ximénez must against p,p'-DDE-induced liver damages in aged Mus spretus mice. Food Chem Toxicol 2020; 136:110984. [DOI: 10.1016/j.fct.2019.110984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
|
37
|
Chen L, Xiang E, Li C, Han B, Zhang Q, Rao W, Xiao C, Wu D. Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorate Nephrocyte Injury and Proteinuria in a Diabetic Nephropathy Rat Model. J Diabetes Res 2020; 2020:8035853. [PMID: 32405507 PMCID: PMC7206880 DOI: 10.1155/2020/8035853] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are shown to alleviate renal injury of diabetic nephropathy (DN) in rats. However, the underlying mechanism of this beneficial effect is not fully understood. The aims of this study are to evaluate effects of umbilical cord-derived mesenchymal stem cells (UC-MSCs) on renal cell apoptosis in streptozotocin- (STZ-) induced diabetic rats and explore the underlying mechanisms. Characteristics of UC-MSCs were identified by flow cytometry and differentiation capability. Six weeks after DN induction by STZ injection in Sprague-Dawley rats, the DN rats received UC-MSCs once a week for consecutive two weeks. DN-related physical and biochemical parameters were measured at 2 weeks after UC-MSC infusion. Renal histological changes were also assessed. Moreover, the apoptosis of renal cells and expression of apoptosis-related proteins were evaluated. Compared with DN rats, rats treated with UC-MSCs showed suppressed increase in 24-hour urinary total protein, urinary albumin to creatinine ratio, serum creatinine, and blood urea nitrogen. UC-MSC treatment ameliorated pathological abnormalities in the kidney of DN rats as evidenced by H&E, PAS, and Masson Trichrome staining. Furthermore, UC-MSC treatment reduced apoptosis of renal cells in DN rats. UC-MSCs promoted expression of antiapoptosis protein Bcl-xl and suppressed expression of high mobility group protein B1 (HMGB1) in the kidney of DN rats. Most importantly, UC-MSCs suppressed upregulation of thioredoxin-interacting protein (TXNIP), downregulation of thioredoxin 1 (TRX1), and activation of apoptosis signal-regulating kinase 1 (ASK1) and P38 MAPK in the kidney of DN rats. Our results suggest that UC-MSCs could alleviate nephrocyte injury and albuminuria of DN rats through their antiapoptotic property. The protective effects of UC-MSCs may be mediated by inhibiting TXNIP upregulation in part.
Collapse
Affiliation(s)
- Lian Chen
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - E. Xiang
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| | - Changyong Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Bing Han
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| | - Wei Rao
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| | - Cuihong Xiao
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, China
| |
Collapse
|
38
|
Wang Y, Wen H, Fu J, Cai L, Li PL, Zhao CL, Dong ZF, Ma JP, Wang X, Tian H, Zhang Y, Liu Y, Cai J, She ZG, Huang Z, Li W, Li H. Hepatocyte TNF Receptor-Associated Factor 6 Aggravates Hepatic Inflammation and Fibrosis by Promoting Lysine 6-Linked Polyubiquitination of Apoptosis Signal-Regulating Kinase 1. Hepatology 2020; 71:93-111. [PMID: 31222801 DOI: 10.1002/hep.30822] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/14/2019] [Indexed: 12/24/2022]
Abstract
Activation of apoptosis signal-regulating kinase 1 (ASK1) is a key driving force of the progression of nonalcoholic steatohepatitis (NASH) and represents an attractive therapeutic target for NASH treatment. However, the molecular and cellular mechanisms underlying ASK1 activation in the pathogenesis of NASH remain incompletely understood. In this study, our data unequivocally indicated that hyperactivated ASK1 in hepatocytes is a potent inducer of hepatic stellate cell (HSC) activation by promoting the production of hepatocyte-derived factors. Our previous serial studies have shown that the ubiquitination system plays a key role in regulating ASK1 activity during NASH progression. Here, we further demonstrated that tumor necrosis factor receptor-associated factor 6 (TRAF6) promotes lysine 6 (Lys6)-linked polyubiquitination and subsequent activation of ASK1 to trigger the release of robust proinflammatory and profibrotic factors in hepatocytes, which, in turn, drive HSC activation and hepatic fibrosis. Consistent with the in vitro findings, diet-induced liver inflammation and fibrosis were substantially attenuated in Traf6+/- mice, whereas hepatic TRAF6 overexpression exacerbated these abnormalities. Mechanistically, Lys6-linked ubiquitination of ASK1 by TRAF6 facilitates the dissociation of thioredoxin from ASK1 and N-terminal dimerization of ASK1, resulting in the boosted activation of ASK1-c-Jun N-terminal kinase 1/2 (JNK1/2)-mitogen-activated protein kinase 14(p38) signaling cascade in hepatocytes. Conclusion: These results suggest that Lys6-linked polyubiquitination of ASK1 by TRAF6 represents a mechanism underlying ASK1 activation in hepatocytes and a key driving force of proinflammatory and profibrogenic responses in NASH. Thus, inhibiting Lys6-linked polyubiquitination of ASK1 may serve as a potential therapeutic target for NASH treatment.
Collapse
Affiliation(s)
- Yutao Wang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Huan Wen
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Jiajun Fu
- Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lin Cai
- Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng-Long Li
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Chang-Ling Zhao
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Zhu-Feng Dong
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Jun-Peng Ma
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xi Wang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Han Tian
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Yan Zhang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Ye Liu
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Jingjing Cai
- Institute of Model Animal of Wuhan University, Wuhan, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhi-Gang She
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Zan Huang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Wenhua Li
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hongliang Li
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
39
|
Bai L, Chen MM, Chen ZD, Zhang P, Tian S, Zhang Y, Zhu XY, Liu Y, She ZG, Ji YX, Li H. F-box/WD Repeat-Containing Protein 5 Mediates the Ubiquitination of Apoptosis Signal-Regulating Kinase 1 and Exacerbates Nonalcoholic Steatohepatitis in Mice. Hepatology 2019; 70:1942-1957. [PMID: 30703849 DOI: 10.1002/hep.30537] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/27/2019] [Indexed: 12/17/2022]
Abstract
Inhibition of apoptosis signal-regulating kinase 1 (ASK1) activation has emerged as a promising target for the treatment of nonalcoholic steatohepatitis (NASH). Multiple forms of posttranslational modifications determine the activity of ASK1. In addition to phosphorylation, recent studies revealed that ubiquitination is essential for ASK1 activation. However, the endogenous factor that regulates ASK1 ubiquitination and activation remains poorly defined. In this study, we identified the E3 ligase Skp1-Cul1-F-box (SCF) protein F-box/WD repeat-containing protein 5 (FBXW5) as a key endogenous activator of ASK1 ubiquitination. FBXW5 is the central component of the SCF complex (SCFFbxw5 ) that directly interacts with and ubiquitinates ASK1 in hepatocytes during NASH development. An in vivo study showed that hepatocyte-specific overexpression of FBXW5 exacerbated diet-induced systemic and hepatic metabolic disorders, as well as the activation of ASK1-related mitogen-activated protein kinase (MAPK) signaling in the liver. Conversely, hepatocyte-specific deletion of FBXW5 significantly prevented the progression of these abnormalities. Mechanically, FBXW5 facilitated the addition of Lys63-linked ubiquitin to ASK1 and thus exacerbated ASK1-c-Jun N-terminal kinase/p38 MAPK signaling, inflammation, and lipid accumulation. Furthermore, we demonstrated that the N-terminus (S1) and C-terminus (S3) of FBXW5 respectively and competitively ablate the function of FBXW5 on ASK1 activation and served as effective inhibitors of NASH progression. Conclusion: This evidence strongly suggests that SCFFbxw5 is an important activator of ASK1 ubiquitination in the context of NASH. The development of FBXW5(S1) or FBXW5(S3)-mimicking drugs and screening of small-molecular inhibitors specifically abrogating ASK1 ubiquitination-dependent activation are viable approaches for NASH treatment.
Collapse
Affiliation(s)
- Lan Bai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ming-Ming Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ze-Dong Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ye Liu
- Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animals of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Huang Z, Shu J, Jiang W, Jiang M, Lu Y, Dai H, Xu N, Yuan H, Cai J. Regulator of G Protein Signaling 6 Facilities Cardiac Hypertrophy by Activating Apoptosis Signal-Regulating Kinase 1-P38/c-JUN N-Terminal Kinase 1/2 Signaling. J Am Heart Assoc 2019; 7:e009179. [PMID: 30371330 PMCID: PMC6404897 DOI: 10.1161/jaha.118.009179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Regulator of G protein signaling 6 (RGS6) is an important member of the RGS family and produces pleiotropic regulatory effects on cardiac pathophysiology. However, the role of RGS6 protein in cardiomyocytes during angiotensin II– and pressure overload–induced cardiac hypertrophy remain unknown. Methods and Results Here, we used a genetic approach to study the regulatory role of RGS6 in cardiomyocytes during pathological cardiac hypertrophy. RGS6 expression was significantly increased in failing human hearts and in hypertrophic murine hearts. The extent of aortic banding–induced cardiac hypertrophy, dysfunction, and fibrosis in cardiac‐specific RGS6 knockout mice was alleviated, whereas the hearts of transgenic mice with cardiac‐specific RGS6 overexpression exhibited exacerbated responses to pressure overload. Consistent with these findings, RGS6 also facilitated an angiotensin II–induced hypertrophic response in isolated cardiomyocytes. According to the mechanistic studies, RGS6 mediated cardiac hypertrophy by directly interacting with apoptosis signal–regulating kinase 1, which further activates the P38‐c‐JUN N‐terminal kinase 1/2 signaling pathway. Conclusions Based on our findings, RGS6 aggravates cardiac hypertrophy, and the RGS6‐apoptosis signal–regulating kinase 1 pathway represents a potential therapeutic target to attenuate pressure overload–driven cardiac remodeling.
Collapse
Affiliation(s)
- Zhijun Huang
- 2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| | - Jingxian Shu
- 2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| | - Weihong Jiang
- 1 Department of Cardiology The Third Xiangya Hospital Central South University Changsha China
| | - Mengqing Jiang
- 1 Department of Cardiology The Third Xiangya Hospital Central South University Changsha China
| | - Yao Lu
- 2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| | - Haijiang Dai
- 1 Department of Cardiology The Third Xiangya Hospital Central South University Changsha China
| | - Nana Xu
- 2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| | - Hong Yuan
- 2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| | - Jingjing Cai
- 1 Department of Cardiology The Third Xiangya Hospital Central South University Changsha China.,2 The Center of Clinical Pharmacology The Third Xiangya Hospital Central South University Changsha China
| |
Collapse
|
41
|
Psenakova K, Hexnerova R, Srb P, Obsilova V, Veverka V, Obsil T. The redox‐active site of thioredoxin is directly involved in apoptosis signal‐regulating kinase 1 binding that is modulated by oxidative stress. FEBS J 2019; 287:1626-1644. [DOI: 10.1111/febs.15101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/10/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Katarina Psenakova
- Department of Physical and Macromolecular Chemistry Faculty of Science Charles University Prague Czech Republic
- Department of Structural Biology of Signaling Proteins, Division BIOCEV Institute of Physiology of the Czech Academy of Sciences Vestec Czech Republic
| | - Rozalie Hexnerova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic
| | - Pavel Srb
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic
| | - Veronika Obsilova
- Department of Structural Biology of Signaling Proteins, Division BIOCEV Institute of Physiology of the Czech Academy of Sciences Vestec Czech Republic
| | - Vaclav Veverka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Prague Czech Republic
- Department of Cell Biology Faculty of Science Charles University Prague Czech Republic
| | - Tomas Obsil
- Department of Physical and Macromolecular Chemistry Faculty of Science Charles University Prague Czech Republic
- Department of Structural Biology of Signaling Proteins, Division BIOCEV Institute of Physiology of the Czech Academy of Sciences Vestec Czech Republic
| |
Collapse
|
42
|
Baig MH, Baker A, Ashraf GM, Dong JJ. ASK1 and its role in cardiovascular and other disorders: available treatments and future prospects. Expert Rev Proteomics 2019; 16:857-870. [DOI: 10.1080/14789450.2019.1676735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Abu Baker
- Nanobiotechnology and nanomedicine lab, Department of Biosciences, Integral University, Lucknow, India
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jae-June Dong
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
43
|
Zhang W, Wang X, Yu M, Li JA, Meng H. The c-Jun N-terminal kinase signaling pathway in epilepsy: activation, regulation, and therapeutics. J Recept Signal Transduct Res 2019; 38:492-498. [PMID: 31038026 DOI: 10.1080/10799893.2019.1590410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epilepsy affects approximately 50-70 million people worldwide and 30-40% of patients do not benefit from medication. Therefore, it is necessary to identify novel targets for epileptic treatments. c-Jun N-terminal kinase (JNK) is a member of the mitogen-activated protein kinase (MAPK) family that activates diverse substrates, such as transcriptional factors, adaptor proteins, and signaling proteins, and has a wide variety of functions in both physiological and pathological conditions. The excessive activation of JNK is found not only in the acute phase of epilepsy, but also in the chronic phase, which potentiates it as a promising target in epilepsy control. In this review, we discuss the activation of the JNK pathway in epilepsy and its role in neuronal death, astrocyte activation, and mossy fiber sprouting (MFS) based on recent updates. Finally, we briefly introduce the current agents that target JNK signaling to control epilepsy.
Collapse
Affiliation(s)
- Wuqiong Zhang
- a Department of Neurology and Neuroscience center , The First Hospital of Jilin University , Changchun , P. R. China
| | - Xue Wang
- a Department of Neurology and Neuroscience center , The First Hospital of Jilin University , Changchun , P. R. China
| | - Miaomiao Yu
- a Department of Neurology and Neuroscience center , The First Hospital of Jilin University , Changchun , P. R. China
| | - Jia-Ai Li
- a Department of Neurology and Neuroscience center , The First Hospital of Jilin University , Changchun , P. R. China
| | - Hongmei Meng
- a Department of Neurology and Neuroscience center , The First Hospital of Jilin University , Changchun , P. R. China
| |
Collapse
|
44
|
Liu S, Shen M, Li C, Wei Y, Meng X, Li R, Cao Y, Wu W, Liu H. PKCδ contributes to oxidative stress-induced apoptosis in porcine ovarian granulosa cells via activating JNK. Theriogenology 2019; 131:89-95. [PMID: 30965208 DOI: 10.1016/j.theriogenology.2019.03.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 01/21/2023]
Abstract
Oxidative stress-induced apoptosis of granulosa cells (GCs) is believed to be an important cause of follicular atresia. Our previous work showed that the c-Jun N-terminal kinase (also known as JNK) might promote apoptosis in GCs during oxidative stress. The aim of this study was to investigate the upstream signaling required for JNK-mediated GCs apoptosis during oxidative stress. Since PKCδ and ASK1 have been suggested to regulate JNK activity in some types of cells, we hypothesized that PKCδ and ASK1 might contribute to JNK-dependent apoptosis in GCs suffering oxidative stimulation. To test this assumption, porcine GCs obtained from healthy follicles were treated with H2O2 alone, or together with inhibitors against PKCδ and JNK, and then collected for cell viability assay, TUNEL staining, immunoprecipitation, western blotting, or JNK activity detection in vitro. The current results showed that the cell viability loss, DNA fragmentation, morphological shrinkage, and nuclear condensation in H2O2-treated porcine GCs was correlated with enhanced activation of JNK. Although ASK1 was supposed to be a JNK activator, we found no definite role of ASK1 in JNK-induced GCs apoptosis during oxidative stress. Further investigations revealed that H2O2-mediated PKCδ activation was required for the apoptotic death of porcine GCs. Particularly, the pro-apoptotic effects of PKCδ on porcine GCs might be achieved by activating the mitochondrial pathway. Importantly, we found that p-PKCδ acts as an upstream activator of JNK in H2O2-treated porcine GCs. However, JNK has no regulatory effect on PKCδ activity. Taken together, our findings provided a novel model of GCs apoptosis involving the activation of PKCδ/JNK/mitochondrial apoptosis axis during oxidative stress.
Collapse
Affiliation(s)
- Shuo Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Chengyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yinghui Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xueqing Meng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Rongyang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yan Cao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
45
|
Sun P, Zeng Q, Cheng D, Zhang K, Zheng J, Liu Y, Yuan YF, Tang YD. Caspase Recruitment Domain Protein 6 Protects Against Hepatic Steatosis and Insulin Resistance by Suppressing Apoptosis Signal-Regulating Kinase 1. Hepatology 2018; 68:2212-2229. [PMID: 29729191 DOI: 10.1002/hep.30075] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/30/2018] [Indexed: 12/25/2022]
Abstract
The rapidly increasing prevalence of metabolic disorders associated with nonalcoholic fatty liver disease (NAFLD) warrants further study of the underlying mechanisms to identify key regulators as targets for the development of therapeutic interventions. Caspase recruitment domain protein 6 (Card6), as a member of the CARD family that regulates cell death and immunity, may potentially control this process. Indeed, Card6 down-regulation was found to be closely associated with the fatty livers observed in NAFLD patients, obese mice, and a palmitate-treated hepatocyte model. Gain-of-function and loss-of-function Card6 mouse models demonstrated that Card6 protected mice from insulin resistance, hepatic steatosis, and inflammatory responses upon high-fat diet administration. Mechanistically, Card6 interacted with and inhibited apoptosis signal-regulating kinase 1 (Ask1) and its subsequent downstream c-Jun N-terminal kinase/p38 signaling. Furthermore, Ask1 was sufficient to mediate Card6 function, and the interaction between Ask1 and Card6 was absolutely required for Card6 function in vivo. Adenovirus-mediated Card6 overexpression in the liver effectively ameliorated insulin resistance and hepatic steatosis in ob/ob mice. Therefore, we identified Card6 as an important negative regulator in NAFLD. Conclusion: Targeting Ask1 by Card6 may be a good strategy to develop a therapeutic method against NAFLD.
Collapse
Affiliation(s)
- Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Zeng
- Health Management Institute, Chinese PLA General Hospital, Beijing, China
| | - Daqing Cheng
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kuo Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jilin Zheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupeng Liu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Feng Yuan
- Department of Hepatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
46
|
Ji N, Yang Y, Cai CY, Lei ZN, Wang JQ, Gupta P, Shukla S, Ambudkar SV, Kong D, Chen ZS. Selonsertib (GS-4997), an ASK1 inhibitor, antagonizes multidrug resistance in ABCB1- and ABCG2-overexpressing cancer cells. Cancer Lett 2018; 440-441:82-93. [PMID: 30315846 DOI: 10.1016/j.canlet.2018.10.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 01/26/2023]
Abstract
Overexpression of ATP-binding cassette (ABC) transporters is one of the most important mechanisms responsible for the development of multidrug resistance (MDR). Selonsertib, a serine/threonine kinase inhibitor, targets apoptosis signal-regulating kinase 1 (ASK1) and is now in phase III clinical trial for the treatment of non-alcoholic steatohepatitis (NASH). In this study, we investigated whether selonsertib could reverse MDR-mediated by ABC transporters, including ABCB1, ABCG2, ABCC1 and ABCC10. The results showed that selonsertib significantly reversed ABCB1- and ABCG2-mediated MDR, but not MDR-mediated by ABCC1 or ABCC10. Mechanism studies indicated that the reversal effect of selonsertib was related to the attenuation of the efflux activity of ABCB1 and ABCG2 transporters, without the protein level decrease or change in the subcellular localization of ABCB1 or ABCG2. Selonsertib stimulated the ATPase activity of ABCB1 and ABCG2 in a concentration-dependent manner, and in silico docking study showed selonsertib could interact with the substrate-binding sites of both ABCB1 and ABCG2. This study provides a clue into a novel treatment strategy, which includes a combination of selonsertib with antineoplastic drugs to attenuate MDR-mediated by ABCB1 or ABCG2 in cancer cells overexpressing these transporters.
Collapse
Affiliation(s)
- Ning Ji
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Suneet Shukla
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, 20892, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, 20892, USA
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
47
|
Abrams SL, Lertpiriyapong K, Yang LV, Martelli AM, Cocco L, Ratti S, Falasca M, Murata RM, Rosalen PL, Lombardi P, Libra M, Candido S, Montalto G, Cervello M, Steelman LS, McCubrey JA. Introduction of WT-TP53 into pancreatic cancer cells alters sensitivity to chemotherapeutic drugs, targeted therapeutics and nutraceuticals. Adv Biol Regul 2018; 69:16-34. [PMID: 29980405 DOI: 10.1016/j.jbior.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 06/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5-10%. Mutations at the TP53 gene are readily detected in pancreatic tumors isolated from PDAC patients. We have investigated the effects of restoration of wild-type (WT) TP53 activity on the sensitivity of pancreatic cancer cells to: chemotherapy, targeted therapy, as well as, nutraceuticals. Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Likewise, the sensitivity to drugs used to treat other cancers such as: doxorubicin, mitoxantrone, and 4 hydroxy tamoxifen (4HT) also increased upon introduction of WT-TP53 into MIA-PaCa-2 cells. Furthermore, the sensitivity to certain inhibitors which target: PI3K/mTORC1, PDK1, SRC, GSK-3, and biochemical processes such as proteasomal degradation and the nutraceutical berberine as increased upon introduction of WT-TP53. Furthermore, in some cases, cells with WT-TP53 were more sensitive to the combination of drugs and suboptimal doses of the MDM2 inhibitor nutlin-3a. However, TP53-independent effects of nutlin-3a were observed upon treatment with either a proteasomal or a PI3K/mTOR inhibitor. These studies indicate the sensitizing effects that WT-TP53 can have in PDAC cells which normally lack WT-TP53 to various therapeutic agents and suggest approaches to improve PDAC therapy.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Ramiro M Murata
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy; Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
48
|
Amos LA, Ma FY, Tesch GH, Liles JT, Breckenridge DG, Nikolic-Paterson DJ, Han Y. ASK1 inhibitor treatment suppresses p38/JNK signalling with reduced kidney inflammation and fibrosis in rat crescentic glomerulonephritis. J Cell Mol Med 2018; 22:4522-4533. [PMID: 29998485 PMCID: PMC6111820 DOI: 10.1111/jcmm.13705] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/29/2018] [Indexed: 12/15/2022] Open
Abstract
Activation of p38 mitogen‐activated protein kinase (MAPK) and c‐Jun amino terminal kinase (JNK) is prominent in human crescentic glomerulonephritis. p38 and JNK inhibitors suppress crescentic disease in animal models; however, the upstream mechanisms inducing activation of these kinases in crescentic glomerulonephritis are unknown. We investigated the hypothesis that apoptosis signal‐regulating kinase 1 (ASK1/MAP3K5) promote p38/JNK activation and renal injury in models of nephrotoxic serum nephritis (NTN); acute glomerular injury in SD rats, and crescentic disease in WKY rats. Treatment with the selective ASK1 inhibitor, GS‐444217 or vehicle began 1 hour before nephrotoxic serum injection and continued until animals were killed on day 1 (SD rats) or 14 (WKY rats). NTN resulted in phosphorylation (activation) of p38 and c‐Jun in both models which was substantially reduced by ASK1 inhibitor treatment. In SD rats, GS‐444217 prevented proteinuria and glomerular thrombosis with suppression of macrophage activation on day 1 NTN. In WKY rats, GS‐444217 reduced crescent formation, prevented renal impairment and reduced proteinuria on day 14 NTN. Macrophage activation, T‐cell infiltration and renal fibrosis were also reduced by GS‐444217. In conclusion, GS‐444217 treatment inhibited p38/JNK activation and development of renal injury in rat NTN. ASK1 inhibitors may have therapeutic potential in rapidly progressive glomerulonephritis.
Collapse
Affiliation(s)
- Liv A Amos
- Department of Nephrology, Monash Medical Centre, Clayton, Vic., 3168, Australia.,Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Vic., 3168, Australia
| | - Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Clayton, Vic., 3168, Australia.,Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Vic., 3168, Australia
| | - Greg H Tesch
- Department of Nephrology, Monash Medical Centre, Clayton, Vic., 3168, Australia.,Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Vic., 3168, Australia
| | | | | | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Clayton, Vic., 3168, Australia.,Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Vic., 3168, Australia
| | - Yingjie Han
- Department of Nephrology, Monash Medical Centre, Clayton, Vic., 3168, Australia.,Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Vic., 3168, Australia
| |
Collapse
|
49
|
Candido S, Abrams SL, Steelman L, Lertpiriyapong K, Martelli AM, Cocco L, Ratti S, Follo MY, Murata RM, Rosalen PL, Lombardi P, Montalto G, Cervello M, Gizak A, Rakus D, Suh PG, Libra M, McCubrey JA. Metformin influences drug sensitivity in pancreatic cancer cells. Adv Biol Regul 2018; 68:13-30. [PMID: 29482945 DOI: 10.1016/j.jbior.2018.02.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 06/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5-10% after diagnosis and treatment. Pancreatic cancer has been associated with type II diabetes as the frequency of recently diagnosed diabetics that develop pancreatic cancer within a 10-year period of initial diagnosis of diabetes in increased in comparison to non-diabetic patients. Metformin is a very frequently prescribed drug used to treat type II diabetes. Metformin acts in part by stimulating AMP-kinase (AMPK) and results in the suppression of mTORC1 activity and the induction of autophagy. In the following studies, we have examined the effects of metformin in the presence of various chemotherapeutic drugs, signal transduction inhibitors and natural products on the growth of three different PDAC lines. Metformin, by itself, was not effective at suppressing growth of the pancreatic cancer cell lines at concentration less than 1000 nM, however, in certain PDAC lines, a suboptimal dose of metformin (250 nM) potentiated the effects of various chemotherapeutic drugs used to treat pancreatic cancer (e.g., gemcitabine, cisplatin, 5-fluorouracil) and other cancer types (e.g., doxorubicin, docetaxel). Furthermore, metformin could increase anti-proliferative effects of mTORC1 and PI3K/mTOR inhibitors as well as natural products such as berberine and the anti-malarial drug chloroquine in certain PDAC lines. Thus, metformin can enhance the effects of certain drugs and signal transduction inhibitors which are used to treat pancreatic and various other cancers.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Linda Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Ramiro M Murata
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Pann-Gill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
50
|
Selvaggio G, Coelho PMBM, Salvador A. Mapping the phenotypic repertoire of the cytoplasmic 2-Cys peroxiredoxin - Thioredoxin system. 1. Understanding commonalities and differences among cell types. Redox Biol 2018; 15:297-315. [PMID: 29304480 PMCID: PMC5975082 DOI: 10.1016/j.redox.2017.12.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022] Open
Abstract
The system (PTTRS) formed by typical 2-Cys peroxiredoxins (Prx), thioredoxin (Trx), Trx reductase (TrxR), and sulfiredoxin (Srx) is central in antioxidant protection and redox signaling in the cytoplasm of eukaryotic cells. Understanding how the PTTRS integrates these functions requires tracing phenotypes to molecular properties, which is non-trivial. Here we analyze this problem based on a model that captures the PTTRS' conserved features. We have mapped the conditions that generate each distinct response to H2O2 supply rates (vsup), and estimated the parameters for thirteen human cell types and for Saccharomyces cerevisiae. The resulting composition-to-phenotype map yielded the following experimentally testable predictions. The PTTRS permits many distinct responses including ultra-sensitivity and hysteresis. However, nearly all tumor cell lines showed a similar response characterized by limited Trx-S- depletion and a substantial but self-limited gradual accumulation of hyperoxidized Prx at high vsup. This similarity ensues from strong correlations between the TrxR, Srx and Prx activities over cell lines, which contribute to maintain the Prx-SS reduction capacity in slight excess over the maximal steady state Prx-SS production. In turn, in erythrocytes, hepatocytes and HepG2 cells high vsup depletes Trx-S- and oxidizes Prx mainly to Prx-SS. In all nucleated human cells the Prx-SS reduction capacity defined a threshold separating two different regimes. At sub-threshold vsup the cytoplasmic H2O2 concentration is determined by Prx, nM-range and spatially localized, whereas at supra-threshold vsup it is determined by much less active alternative sinks and μM-range throughout the cytoplasm. The yeast shows a distinct response where the Prx Tsa1 accumulates in sulfenate form at high vsup. This is mainly due to an exceptional stability of Tsa1's sulfenate. The implications of these findings for thiol redox regulation and cell physiology are discussed. All estimates were thoroughly documented and provided, together with analytical approximations for system properties, as a resource for quantitative redox biology.
Collapse
Affiliation(s)
- Gianluca Selvaggio
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; MIT-Portugal Program Bioengineering Systems Doctoral Program, Portugal
| | - Pedro M B M Coelho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Armindo Salvador
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CQC, Department of Chemistry, University of Coimbra, Portugal.
| |
Collapse
|