1
|
Ashok A, Ashwathnarayan A, Bhaskar S, Shekar S, Kalathur G, Prasanna J, Kumar A. Inhibition of proteasome activity facilitates definitive endodermal specification of pluripotent stem cells by influencing YAP signalling. Life Sci 2024; 358:123160. [PMID: 39433087 DOI: 10.1016/j.lfs.2024.123160] [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: 05/27/2024] [Revised: 08/03/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
AIMS The knowledge of the molecular players that regulate the generation of endoderm cells is imperative to obtain homogenous population of pancreatic β-cells from stem cells. The Ubiquitin proteasome system (UPS) has been envisaged as a crucial intracellular protein degradation system, but its role in the generation of β-cells remains elusive. Hence, it would be appropriate to unravel the potential role of UPS in endoderm specification and utilize the understanding to generate β-cells from pluripotent stem cells. MATERIALS AND METHODS The pluripotent stem cells (mESCs, miPSCs and hIPSCs) were subjected to differentiation towards pancreatic β-cells and assessed the proteasomal activity during endodermal differentiation. Pharmacologic agents MG132 and IU-1 were employed to inhibit and activate proteasomal activity respectively at the definitive endoderm stage to investigate its impact on the generation of β-cells. The expression of stage-specific genes were analyzed at transcript and protein levels. We also explored the role of unfolded protein response and UPS-regulated signalling pathways in endodermal differentiation. KEY FINDINGS We observed decreased proteasomal activity specifically during endoderm, but not during the generation of other lineages. Extraneous proteasomal inhibition enhanced the expression of endodermal genes while increasing the proteasomal activity hindered definitive endodermal differentiation. Proteasomal inhibition at the definitive endodermal stage culminated in an enriched generation of insulin-positive cells. Elevated endodermal gene expression was consistent in mESCs and hIPSCs upon proteasomal inhibition. Mechanistic insight revealed the proteasome-inhibited enhanced endodermal differentiation to be via modulating the YAP pathway. SIGNIFICANCE Our study unravels the specific involvement of UPS in endoderm cell generation from pluripotent stem cells and paves the way for obtaining potential definitive endodermal cells for plausible cellular therapy in the future.
Collapse
Affiliation(s)
- Akshaya Ashok
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Ashwini Ashwathnarayan
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Spandana Shekar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Guruprasad Kalathur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Ashok A, Kalthur G, Kumar A. Degradation meets development: Implications in β-cell development and diabetes. Cell Biol Int 2024; 48:759-776. [PMID: 38499517 DOI: 10.1002/cbin.12155] [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: 11/03/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Pancreatic development is orchestrated by timely synthesis and degradation of stage-specific transcription factors (TFs). The transition from one stage to another stage is dependent on the precise expression of the developmentally relevant TFs. Persistent expression of particular TF would impede the exit from the progenitor stage to the matured cell type. Intracellular protein degradation-mediated protein turnover contributes to a major extent to the turnover of these TFs and thereby dictates the development of different tissues. Since even subtle changes in the crucial cellular pathways would dramatically impact pancreatic β-cell performance, it is generally acknowledged that the biological activity of these pathways is tightly regulated by protein synthesis and degradation process. Intracellular protein degradation is executed majorly by the ubiquitin proteasome system (UPS) and Lysosomal degradation pathway. As more than 90% of the TFs are targeted to proteasomal degradation, this review aims to examine the crucial role of UPS in normal pancreatic β-cell development and how dysfunction of these pathways manifests in metabolic syndromes such as diabetes. Such understanding would facilitate designing a faithful approach to obtain a therapeutic quality of β-cells from stem cells.
Collapse
Affiliation(s)
- Akshaya Ashok
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Guruprasad Kalthur
- Division of Reproductive and Developmental Biology, Department of Reproductive Science, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
3
|
Yin Y, Tan M, Han L, Zhang L, Zhang Y, Zhang J, Pan W, Bai J, Jiang T, Li H. The hippo kinases MST1/2 in cardiovascular and metabolic diseases: A promising therapeutic target option for pharmacotherapy. Acta Pharm Sin B 2023; 13:1956-1975. [PMID: 37250161 PMCID: PMC10213817 DOI: 10.1016/j.apsb.2023.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/09/2022] [Accepted: 11/18/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders are major components of noncommunicable diseases, causing an enormous health and economic burden worldwide. There are common risk factors and developmental mechanisms among them, indicating the far-reaching significance in exploring the corresponding therapeutic targets. MST1/2 kinases are well-established proapoptotic effectors that also bidirectionally regulate autophagic activity. Recent studies have demonstrated that MST1/2 influence the outcome of cardiovascular and metabolic diseases by regulating immune inflammation. In addition, drug development against them is in full swing. In this review, we mainly describe the roles and mechanisms of MST1/2 in apoptosis and autophagy in cardiovascular and metabolic events as well as emphasis on the existing evidence for their involvement in immune inflammation. Moreover, we summarize the latest progress of pharmacotherapy targeting MST1/2 and propose a new mode of drug combination therapy, which may be beneficial to seek more effective strategies to prevent and treat CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yunfei Yin
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Mingyue Tan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lianhua Han
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lei Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yue Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jun Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wanqian Pan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaxiang Bai
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tingbo Jiang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongxia Li
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
4
|
Uhlemeyer C, Müller N, Rieck M, Kuboth J, Schlegel C, Grieß K, Dorweiler TF, Heiduschka S, Eckel J, Roden M, Lammert E, Stoffel M, Belgardt BF. Selective ablation of P53 in pancreatic beta cells fails to ameliorate glucose metabolism in genetic, dietary and pharmacological models of diabetes mellitus. Mol Metab 2022; 67:101650. [PMID: 36470401 PMCID: PMC9791454 DOI: 10.1016/j.molmet.2022.101650] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Beta cell dysfunction and death are critical steps in the development of both type 1 and type 2 diabetes (T1D and T2D), but the underlying mechanisms are incompletely understood. Activation of the essential tumor suppressor and transcription factor P53 (also known as TP53 and Trp53 in mice) was linked to beta cell death in vitro and has been reported in several diabetes mouse models and beta cells of humans with T2D. In this article, we set out to determine the beta cell specific role of P53 in beta cell dysfunction, cell death and development of diabetes in vivo. METHODS We generated beta cell specific P53 knockout (P53BKO) mice and used complementary genetic, dietary and pharmacological models of glucose intolerance, beta cell dysfunction and diabetes development to evaluate the functional role of P53 selectively in beta cells. We further analyzed the effect of P53 ablation on beta cell survival in isolated pancreatic islets exposed to diabetogenic stress inducers ex vivo by flow cytometry. RESULTS Beta cell specific ablation of P53/Trp53 failed to ameliorate glucose tolerance, insulin secretion or to increase beta cell numbers in genetic, dietary and pharmacological models of diabetes. Additionally, loss of P53 in beta cells did not protect against streptozotocin (STZ) induced hyperglycemia and beta cell death, although STZ-induced activation of classical pro-apoptotic P53 target genes was significantly reduced in P53BKO mice. In contrast, Olaparib mediated PARP1 inhibition protected against acute ex vivo STZ-induced beta cell death and islet destruction. CONCLUSIONS Our study reveals that ablation of P53 specifically in beta cells is unexpectedly unable to attenuate beta cell failure and death in vivo and ex vivo. While during development and progression of diabetes, P53 and P53-regulated pathways are activated, our study suggests that P53 signaling is not essential for loss of beta cells or beta cell dysfunction. P53 in other cell types and organs may predominantly regulate systemic glucose homeostasis.
Collapse
Affiliation(s)
- Celina Uhlemeyer
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| | - Nadine Müller
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michael Rieck
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Jennifer Kuboth
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Caroline Schlegel
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Kerstin Grieß
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Tim Florian Dorweiler
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Sonja Heiduschka
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Jürgen Eckel
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes, Düsseldorf, Germany,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Eckhard Lammert
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany,Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Markus Stoffel
- Institute of Molecular Health Sciences (IMHS), ETH Zürich, Zürich, Switzerland; Competence Center Personalized Medicine, ETH Zürich, Zürich, Switzerland; Medical Faculty, University of Zürich, Zürich, Switzerland
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
| |
Collapse
|
5
|
Field JT, Gordon JW. BNIP3 and Nix: Atypical regulators of cell fate. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119325. [PMID: 35863652 DOI: 10.1016/j.bbamcr.2022.119325] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 11/27/2022]
Abstract
Since their discovery nearly 25 years ago, the BCL-2 family members BNIP3 and BNIP3L (aka Nix) have been labelled 'atypical'. Originally, this was because BNIP3 and Nix have divergent BH3 domains compared to other BCL-2 proteins. In addition, this atypical BH3 domain is dispensable for inducing cell death, which is also unusual for a 'death gene'. Instead, BNIP3 and Nix utilize a transmembrane domain, which allows for dimerization and insertion into and through organelle membranes to elicit cell death. Much has been learned regarding the biological function of these two atypical death genes, including their role in metabolic stress, where BNIP3 is responsive to hypoxia, while Nix responds variably to hypoxia and is also down-stream of PKC signaling and lipotoxic stress. Interestingly, both BNIP3 and Nix respond to signals related to cell atrophy. In addition, our current view of regulated cell death has expanded to include forms of necrosis such as necroptosis, pyroptosis, ferroptosis, and permeability transition-mediated cell death where BNIP3 and Nix have been shown to play context- and cell-type specific roles. Perhaps the most intriguing discoveries in recent years are the results demonstrating roles for BNIP3 and Nix outside of the purview of death genes, such as regulation of proliferation, differentiation/maturation, mitochondrial dynamics, macro- and selective-autophagy. We provide a historical and unbiased overview of these 'death genes', including new information related to alternative splicing and post-translational modification. In addition, we propose to redefine these two atypical members of the BCL-2 family as versatile regulators of cell fate.
Collapse
Affiliation(s)
- Jared T Field
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Canada; The Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Joseph W Gordon
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Canada; College of Nursing, Rady Faculty of Health Science, University of Manitoba, Canada; The Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
| |
Collapse
|
6
|
Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, Wu H, Gong G. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis 2022; 13:444. [PMID: 35534453 PMCID: PMC9085840 DOI: 10.1038/s41419-022-04906-6] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic organelles that participate in ATP generation and involve calcium homeostasis, oxidative stress response, and apoptosis. Dysfunctional or damaged mitochondria could cause serious consequences even lead to cell death. Therefore, maintaining the homeostasis of mitochondria is critical for cellular functions. Mitophagy is a process of selectively degrading damaged mitochondria under mitochondrial toxicity conditions, which plays an essential role in mitochondrial quality control. The abnormal mitophagy that aggravates mitochondrial dysfunction is closely related to the pathogenesis of many diseases. As the myocardium is a highly oxidative metabolic tissue, mitochondria play a central role in maintaining optimal performance of the heart. Dysfunctional mitochondria accumulation is involved in the pathophysiology of cardiovascular diseases, such as myocardial infarction, cardiomyopathy and heart failure. This review discusses the most recent progress on mitophagy and its role in cardiovascular disease.
Collapse
Affiliation(s)
- Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hanyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Huayan Wu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
7
|
Li Y, Zheng W, Lu Y, Zheng Y, Pan L, Wu X, Yuan Y, Shen Z, Ma S, Zhang X, Wu J, Chen Z, Zhang X. BNIP3L/NIX-mediated mitophagy: molecular mechanisms and implications for human disease. Cell Death Dis 2021; 13:14. [PMID: 34930907 PMCID: PMC8688453 DOI: 10.1038/s41419-021-04469-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/26/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023]
Abstract
Mitophagy is a highly conserved cellular process that maintains the mitochondrial quantity by eliminating dysfunctional or superfluous mitochondria through autophagy machinery. The mitochondrial outer membrane protein BNIP3L/Nix serves as a mitophagy receptor by recognizing autophagosomes. BNIP3L is initially known to clear the mitochondria during the development of reticulocytes. Recent studies indicated it also engages in a variety of physiological and pathological processes. In this review, we provide an overview of how BNIP3L induces mitophagy and discuss the biological functions of BNIP3L and its regulation at the molecular level. We further discuss current evidence indicating the involvement of BNIP3L-mediated mitophagy in human disease, particularly in cancer and neurological disorders.
Collapse
Affiliation(s)
- Yue Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Wanqing Zheng
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Yangyang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Yanrong Zheng
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmacology Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ling Pan
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Xiaoli Wu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Yang Yuan
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Zhe Shen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Shijia Ma
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Xingxian Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Jiaying Wu
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmacology Science, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of The Ministry of Health of China, Zhejiang University, Hangzhou, China.
| |
Collapse
|
8
|
Junaid R, Wahid M, Waseem FS, Habib R, Hasan A. Effect of glucose mediated oxidative stress on apoptotic gene expression in gingival mesenchymal stem cells. BMC Oral Health 2021; 21:653. [PMID: 34922513 PMCID: PMC8684132 DOI: 10.1186/s12903-021-02007-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/01/2021] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Diabetes is a common disease that causes gingival and periodontal problems. Stem cells isolated from dental sources are an emerging area of research with a potential to facilitate regenerative medicine. The stem cells retain the property of self-renewal and the ones isolated from dental sources are mainly multipotent mesenchymal stem cells that have the ability to self-renew as well as differentiation towards multiple lineages.
Objectives
We aimed to isolate and characterize gingival mesenchymal stem cells by pluripotency markers and investigated the effect of oxidative stress on growth kinetics and apoptotic gene expression of gingival cells exposed to glucose mediated oxidative stress.
Methods
In this study, we isolated gingival mesenchymal stem cells from gingiva. This was followed by morphologic analysis using inverted phase contrast microscopy and molecular profiling of these cells for the mRNA expression of specific genes. The isolated cells were cultured till passage 3 and then exposed to oxidative stress (high glucose concentration). We measured the apoptotic gene expression and compared their growth kinetics.
Results
The results showed that oxidative stress produced by glucose reduced growth kinetics and increased apoptotic gene expression in gingival mesenchymal stem cells. According to the genetic results, glucose activated TNF family to initiate apoptosis.
Conclusion
In conclusion, the present study demonstrated that high glucose obliterated cellular proliferation testified by evaluating growth kinetics and induced apoptotic gene expression in gingival mesenchymal stem cells. This initiated extrinsic apoptotic pathway mediated by TNF family. Therefore, in diabetes oral health condition is compromised and periodontal disease is common.
Collapse
|
9
|
Pearson GL, Gingerich MA, Walker EM, Biden TJ, Soleimanpour SA. A Selective Look at Autophagy in Pancreatic β-Cells. Diabetes 2021; 70:1229-1241. [PMID: 34016598 PMCID: PMC8275885 DOI: 10.2337/dbi20-0014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/22/2021] [Indexed: 12/15/2022]
Abstract
Insulin-producing pancreatic β-cells are central to glucose homeostasis, and their failure is a principal driver of diabetes development. To preserve optimal health β-cells must withstand both intrinsic and extrinsic stressors, ranging from inflammation to increased peripheral insulin demand, in addition to maintaining insulin biosynthesis and secretory machinery. Autophagy is increasingly being appreciated as a critical β-cell quality control system vital for glycemic control. Here we focus on the underappreciated, yet crucial, roles for selective and organelle-specific forms of autophagy as mediators of β-cell health. We examine the unique molecular players underlying each distinct form of autophagy in β-cells, including selective autophagy of mitochondria, insulin granules, lipid, intracellular amyloid aggregates, endoplasmic reticulum, and peroxisomes. We also describe how defects in selective autophagy pathways contribute to the development of diabetes. As all forms of autophagy are not the same, a refined view of β-cell selective autophagy may inform new approaches to defend against the various insults leading to β-cell failure in diabetes.
Collapse
Affiliation(s)
- Gemma L Pearson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | | - Emily M Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Veterans Affairs Ann Arbor Health Care System, Ann Arbor, MI
| |
Collapse
|
10
|
Liu J, Lang G, Shi J. Epigenetic Regulation of PDX-1 in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2021; 14:431-442. [PMID: 33564250 PMCID: PMC7866918 DOI: 10.2147/dmso.s291932] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/16/2021] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease characterized by hyperglycemia which is caused by insufficient insulin secretion or insulin resistance. Interaction of genetic, epigenetic and environmental factors plays a significant role in the development of T2DM. Several environmental factors including diet and lifestyle, as well as age have been associated with an increased risk for T2DM. It has been demonstrated that these environmental factors may affect global epigenetic status, and alter the expression of susceptible genes, thereby contributing to the pathogenesis of T2DM. In recent years, a growing body of molecular and genetic studies in diabetes have been focused on the ways to restore the numbers or function of β-cells in order to reverse a range of metabolic consequences of insulin deficiency. The pancreatic duodenal homeobox 1 (PDX-1) is a transcriptional factor that is essential for the development and function of islet cells. A number of studies have shown that there is a significant increase in the level of DNA methylation of PDX-1 resulting in reduced activity in T2DM islets. The decrease in PDX-1 activity may be a critical mediator causing dysregulation of pancreatic β cells in T2DM. This article reviews the epigenetic mechanisms of PDX-1 involved in T2DM, focusing on diabetes and DNA methylation, and discusses some potential strategies for the application of PDX-1 in the treatment of diabetes.
Collapse
Affiliation(s)
- Jiangman Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Guangping Lang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Correspondence: Jingshan Shi Tel +86-851-286-436-66Fax +86-851-286-423-03 Email
| |
Collapse
|
11
|
Bernard H, Teijeiro A, Chaves-Pérez A, Perna C, Satish B, Novials A, Wang JP, Djouder N. Coxsackievirus B Type 4 Infection in β Cells Downregulates the Chaperone Prefoldin URI to Induce a MODY4-like Diabetes via Pdx1 Silencing. CELL REPORTS MEDICINE 2020; 1:100125. [PMID: 33205075 PMCID: PMC7659558 DOI: 10.1016/j.xcrm.2020.100125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/06/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022]
Abstract
Enteroviruses are suspected to contribute to insulin-producing β cell loss and hyperglycemia-induced diabetes. However, mechanisms are not fully defined. Here, we show that coxsackievirus B type 4 (CVB4) infection in human islet-engrafted mice and in rat insulinoma cells displays loss of unconventional prefoldin RPB5 interactor (URI) and PDX1, affecting β cell function and identity. Genetic URI ablation in the mouse pancreas causes PDX1 depletion in β cells. Importantly, diabetic PDX1 heterozygous mice overexpressing URI in β cells are more glucose tolerant. Mechanistically, URI loss triggers estrogen receptor nuclear translocation leading to DNA methyltransferase 1 (DNMT1) expression, which induces Pdx1 promoter hypermethylation and silencing. Consequently, demethylating agent procainamide-mediated DNMT1 inhibition reinstates PDX1 expression and protects against diabetes in pancreatic URI-depleted mice . Finally, the β cells of human diabetes patients show correlations between viral protein 1 and URI, PDX1, and DNMT1 levels. URI and DNMT1 expression and PDX1 silencing provide a causal link between enterovirus infection and diabetes. Coxsackievirus B type 4 infection downregulates URI and affects β cell function Genetic URI ablation in mouse pancreas recapitulates diabetes URI controls Pdx1 methylation via ERα-activating DNMT1 Coxsackievirus B type 4, URI, PDX1, and DNMT1 expression correlate in human pancreata
Collapse
MESH Headings
- Animals
- Capsid Proteins/genetics
- Capsid Proteins/metabolism
- Coxsackievirus Infections/genetics
- Coxsackievirus Infections/metabolism
- Coxsackievirus Infections/pathology
- Coxsackievirus Infections/virology
- DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors
- DNA (Cytosine-5-)-Methyltransferase 1/genetics
- DNA (Cytosine-5-)-Methyltransferase 1/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/virology
- Disease Models, Animal
- Enterovirus B, Human/genetics
- Enterovirus B, Human/metabolism
- Enterovirus B, Human/pathogenicity
- Female
- Gene Expression Regulation
- Glucose/metabolism
- Glucose/pharmacology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/transplantation
- Male
- Mice
- Mice, Transgenic
- Procainamide/pharmacology
- Rats
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Hugo Bernard
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Ana Teijeiro
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Almudena Chaves-Pérez
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
| | - Cristian Perna
- Department of Pathology, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid 28034, Spain
| | - Basanthi Satish
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna Novials
- IDIBAPS, August Pi i Sunyer Biomedical Research Institute and, CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Barcelona, Spain
| | - Jennifer P. Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nabil Djouder
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid 28029, Spain
- Corresponding author
| |
Collapse
|
12
|
Uhlemeyer C, Müller N, Grieß K, Wessel C, Schlegel C, Kuboth J, Belgardt BF. ATM and P53 differentially regulate pancreatic beta cell survival in Ins1E cells. PLoS One 2020; 15:e0237669. [PMID: 32810137 PMCID: PMC7437460 DOI: 10.1371/journal.pone.0237669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/30/2020] [Indexed: 01/09/2023] Open
Abstract
Pancreatic beta cell death is a hallmark of type 1 and 2 diabetes (T1D/T2D), but the underlying molecular mechanisms are incompletely understood. Key proteins of the DNA damage response (DDR), including tumor protein P53 (P53, also known as TP53 or TRP53 in rodents) and Ataxia Telangiectasia Mutated (ATM), a kinase known to act upstream of P53, have been associated with T2D. Here we test and compare the effect of ATM and P53 ablation on beta cell survival in the rat beta cell line Ins1E. We demonstrate that ATM and P53 differentially regulate beta cell apoptosis induced upon fundamentally different types of diabetogenic beta cell stress, including DNA damage, inflammation, lipotoxicity and endoplasmic reticulum (ER) stress. DNA damage induced apoptosis by treatment with the commonly used diabetogenic agent streptozotocin (STZ) is regulated by both ATM and P53. We show that ATM is a key STZ induced activator of P53 and that amelioration of STZ induced cell death by inhibition of ATM mainly depends on P53. While both P53 and ATM control lipotoxic beta cell apoptosis, ATM but not P53 fails to alter inflammatory beta cell death. In contrast, tunicamycin induced (ER stress associated) apoptosis is further increased by ATM knockdown or inhibition, but not by P53 knockdown. Our results reveal differential roles for P53 and ATM in beta cell survival in vitro in the context of four key pathophysiological types of diabetogenic beta cell stress, and indicate that ATM can use P53 independent signaling pathways to modify beta cell survival, dependent on the cellular insult.
Collapse
Affiliation(s)
- Celina Uhlemeyer
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Nadine Müller
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Kerstin Grieß
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Corinna Wessel
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Caroline Schlegel
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Jennifer Kuboth
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- * E-mail:
| |
Collapse
|
13
|
Sun J, Mao L, Yang H, Ren D. Critical role for the Tsc1-mTORC1 pathway in β-cell mass in Pdx1-deficient mice. J Endocrinol 2018; 238:151-163. [PMID: 29875165 PMCID: PMC6030447 DOI: 10.1530/joe-18-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/05/2018] [Indexed: 01/25/2023]
Abstract
Mutations in the pancreatic duodenal homeobox (PDX1) gene are associated with diabetes in humans. Pdx1-haploinsufficient mice also develop diabetes, but the molecular mechanism is unknown. To this end, we knocked down Pdx1 gene expression in mouse MIN6 insulinoma cells. Pdx1 suppression not only increased apoptotic cell death but also decreased cell proliferation, which was associated with a decrease in activity of mechanistic target of rapamycin complex 1 (mTORC1). We found that in Pdx1-deficient mice, tuberous sclerosis 1 (Tsc1) ablation in pancreatic β-cells restores β-cell mass, increases β-cell proliferation and size, decreases the number of TUNEL-positive cells and restores glucose tolerance after glucose challenge. In addition, Tsc1 ablation in pancreatic β-cells increases phosphorylation of initiation factor 4E-binding protein 1 (4E-BP1) phosphorylation and 40S ribosomal protein S6, two downstream targets of mTORC1 indicating that Tsc1 mediates mTORC1 downregulation induced by Pdx1 suppression. These results suggest that the Tsc1-mTORC1 pathway plays an important role in mediating the decrease in β-cell proliferation and growth and the reduction in β-cell mass that occurs in Pdx1-deficient diabetes. Thus, mTORC1 may be target for therapeutic interventions in diabetes associated with reductions in β-cell mass.
Collapse
Affiliation(s)
- Juan Sun
- Department of MedicineThe University of Chicago, Chicago, Illinois, USA
| | - Liqun Mao
- Department of MedicineThe University of Chicago, Chicago, Illinois, USA
| | - Hongyan Yang
- Department of GynecologyKey Research Laboratory of Gynecology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Decheng Ren
- Department of MedicineThe University of Chicago, Chicago, Illinois, USA
- Department of GynecologyKey Research Laboratory of Gynecology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Altered expression of PGC-1α and PDX1 and their methylation status are associated with fetal glucose metabolism in gestational diabetes mellitus. Biochem Biophys Res Commun 2018; 501:300-306. [PMID: 29730292 DOI: 10.1016/j.bbrc.2018.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 11/23/2022]
Abstract
PURPOSE To investigate the effect of gestational diabetes mellitus (GDM) on the expression and methylation of PGC-1α and PDX1 in placenta and their effects on fetal glucose metabolism. METHODS 20 cases of full-term placenta without pregnancy complications and umbilical cord abnormalities and 20 cases of GDM group were collected. DNA and RNA were isolated from samples of tissue collected from the fetal side of the placenta immediately after delivery. DNA methylation was quantified at 7 CpG sites within the PGC-1α and PDX1 genes using PCR amplification of bisulfite treated DNA and subsequent DNA sequencing. PGC-1α and PDX1 mRNA levels were measured by reverse transcription-quantitative PCR (RT-qPCR). Meanwhile, the placental insulin, blood glucose and HbA1c levels were determined. RESULTS The fetus birth weight and placental weight in GDM group were significantly higher than those in control group (P < 0.05). Insulin, HbA1c and blood glucose levels in GDM group were significantly higher than those in control group (P < 0.01). Insulin content was positively correlated with newborn birth weight and placental weight while HbA1c and blood glucose were positively correlated with insulin concentration (r = 0.92, P < 0.01, r = 0.85, P < 0.01). The levels of PGC-1α and PDX1 mRNA were lower in the GDM group compared to the control group. The methylation level of PGC-1α gene was higher in the GDM group compared to the control group (P < 0.05). Blood glucose was negatively correlated with the expression of PGC-1α and PDX1 mRNA in the placenta (r = -0.42, P < 0.01, r = -0.49, P < 0.01). CONCLUSION The changes of epigenetic modification of PGC-1α gene in pregnant women with gestational diabetes mellitus may be a mechanism of abnormal glucose metabolism in offspring.
Collapse
|
15
|
Teucrium polium extract reverses symptoms of streptozotocin-induced diabetes in rats via rebalancing the Pdx1 and FoxO1 expressions. Biomed Pharmacother 2017; 93:1033-1039. [PMID: 28738518 DOI: 10.1016/j.biopha.2017.06.082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/30/2017] [Accepted: 06/20/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Teucrium polium Lamiaceae. an antidiabetic traditional medicine was observed to stimulate insulin secretion and β-cell mass restoration in pancreatic of diabetic rats. Bioactive compounds in T. polium such as terpenoids and flavonoids have a wide range of antioxidant properties. Given this background, our research was aimed at probing the effective molecular mechanism of T. polium which in turn protects the pancreatic β-cells of diabetic rats through attenuation of streptozotocin-induced oxidative stress. MATERIALS AND METHODS Induction of diabetes by streptozotocin (STZ) injection followed by treatment of the rats with T. polium extract (0.5g/kg) and glibenclamide as a standard drug (600μg/kg) for six consecutive weeks. Besides, biochemical indexes including sera glucose and lipids were investigated once every two weeks. Moreover, oxidative stress markers were also measured in the sixth week of treatment to assess the antioxidant capacity of T. polium. Prior to sacrifice of rats, the oral glucose tolerance test was carried out and thereafter western blot analysis was performed on pancreatic tissues to detect the expression of JNK, FoxO1 and Pdx1 proteins in the experimental groups. RESULTS Oral administration of T. polium extract not only disclosed the significant anti-hyperglycemic potential of the plant, but also relieved dyslipidemia and oxidative stress related to diabetes. Furthermore, remarkable improvement of glucose tolerance was exhibited among the treated groups, confirming the presence of insulin in the blood. Subsequently, the effective molecular mechanism of T. polium was partially revealed by western blot analyses which detected considerable up-regulation of p-FoxO1 and Pdx1 proteins parallel to reduction of p-JNK expression among the treated diabetic rats. CONCLUSION This study demonstrates that T. polium extract is able to restore the β-cell mass and insulin secretion by regulation of pivotal transcription factor of the pancreatic β-cells Pdx1 in JNK pathway.
Collapse
|
16
|
Ardestani A, Maedler K. MST1: a promising therapeutic target to restore functional beta cell mass in diabetes. Diabetologia 2016; 59:1843-9. [PMID: 27053234 DOI: 10.1007/s00125-016-3892-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/23/2015] [Indexed: 12/31/2022]
Abstract
The loss of insulin-producing beta cells by apoptosis is a hallmark of all forms of diabetes mellitus. Strategies to prevent beta cell apoptosis and dysfunction are urgently needed to restore the insulin-producing cells and to prevent severe diabetes progression. We recently identified the serine/threonine kinase known as mammalian sterile 20-like kinase 1 (MST1) as a critical regulator of apoptotic beta cell death and dysfunction. MST1 activates several apoptotic signalling pathways, which further stimulate its own cleavage, leading to a vicious cycle of cell death. This led us to hypothesise that MST1 signalling is central to the initiation of beta cell death in diabetes. We found that MST1 is strongly activated in a diabetic beta cell and induces not only its death but also directly impairs insulin secretion through promoting proteasomal degradation of key beta cell transcription factor, pancreatic and duodenal homeobox 1 (PDX1), which is critical for insulin production.Pre-clinical studies in various animal models of diabetes have reported that MST1 deficiency remarkably restores normoglycaemia and beta cell function and prevents the development of diabetes. Importantly, MST1 deficiency can revert fully diabetic beta cells to a non-diabetic state. MST1 may serve as a target for the development of novel therapies for diabetes that trigger the cause of the disease, namely, the destruction of the beta cells. The major current focus of our investigation is to identify and test the efficacy of potent inhibitors of this death signalling pathway to protect beta cells against the effects of autoimmune attack in type 1 diabetes and to preserve beta cell mass and function in type 2 diabetes. This review summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 , and by Harry Heimberg and colleagues, DOI: 10.1007/s00125-016-3879-6 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).
Collapse
Affiliation(s)
- Amin Ardestani
- Islet Biology Laboratory, Centre for Biomolecular Interactions Bremen, University of Bremen, Leobener Straße NW2, Room B2080, 28359, Bremen, Germany.
| | - Kathrin Maedler
- Islet Biology Laboratory, Centre for Biomolecular Interactions Bremen, University of Bremen, Leobener Straße NW2, Room B2080, 28359, Bremen, Germany.
| |
Collapse
|
17
|
Zhang H, Liu J, Wang X, Duan C, Wang X, Yang H. V63 and N65 of overexpressed α-synuclein are involved in mitochondrial dysfunction. Brain Res 2016; 1642:308-318. [PMID: 27048753 DOI: 10.1016/j.brainres.2016.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 01/11/2023]
Abstract
Parkinson's Disease (PD) is one of the most common neurodegenerative diseases. α-Synuclein (α-Syn)-encoded by SNCA, the first-identified PD-related gene-is the main component of Lewy bodies, which are a pathological hallmark of PD. We previously reported that α-Syn accumulates in mitochondria in PD, causing mitochondrial abnormalities and disrupting mitochondrial membrane potential (Δψm) and mitochondrial potential transition pore (mPTP) opening by interacting with the voltage-dependent anion channel (VDAC) and adenine nucleotide translocator. However, the mechanistic basis of mitochondrial impairment caused by α-Syn has yet to be elucidated. It has been suggested that the amino acid residues Q62, V63, and N65 of α-Syn are important for the interaction of the protein with membranes. To investigate whether this underlies the mitochondrial dysfunction induced by α-Syn overexpression, we mutated these residues to alanine and transfected HEK293T and MN9D cells with the mutated forms of α-Syn protein. The V63A and N65A mutations prevented mitochondrial Ca(2+) overload and Δψm dysregulation as well as complex I inactivation and reactive oxygen species production while blocking mPTP opening and caspase 9 activation, possibly by reducing α-Syn accumulation in mitochondria. These results indicate that V63 and N65 are critical residues mediating mitochondrial inactivation. These findings provide novel insight into the molecular events contributing to PD pathogenesis.
Collapse
Affiliation(s)
- Huilin Zhang
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Jia Liu
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xue Wang
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Chunli Duan
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xiaomin Wang
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Hui Yang
- Center of Parkinson's Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of, Education, Department of Neurobiology Capital Medical University, Beijing 100069, China.
| |
Collapse
|
18
|
Ko F, Abadir P, Marx R, Westbrook R, Cooke C, Yang H, Walston J. Impaired mitochondrial degradation by autophagy in the skeletal muscle of the aged female interleukin 10 null mouse. Exp Gerontol 2015; 73:23-7. [PMID: 26596403 DOI: 10.1016/j.exger.2015.11.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/09/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
Mitochondrial dysfunction, chronic inflammation and muscle aging are closely linked. Mitochondrial clearance is a process to dampen inflammation and is a critical pre-requisite to mitobiogenesis. The combined effect of aging and chronic inflammation on mitochondrial degradation by autophagy is understudied. In interleukin 10 null mouse (IL-10(tm/tm)), a rodent model of chronic inflammation, we studied the effects of aging and inflammation on mitochondrial clearance. We show that aging in IL-10(tm/tm) is associated with reduced skeletal muscle mitochondrial death signaling and altered formation of autophagosomes, compared to age-matched C57BL/6 controls. Moreover, skeletal muscles of old IL-10(tm/tm) mice have the highest levels of damaged mitochondria with disrupted mitochondrial ultrastructure and autophagosomes compared to all other groups. These observations highlight the interface between chronic inflammation and aging on altered mitochondrial biology in skeletal muscles.
Collapse
Affiliation(s)
- Fred Ko
- Icahn School of Medicine at Mount Sinai, Brookdale Department of Geriatrics and Palliative Medicine, USA.
| | - Peter Abadir
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, USA
| | - Ruth Marx
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, USA
| | - Reyhan Westbrook
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, USA
| | - Carol Cooke
- Johns Hopkins University, Cellular and Molecular Medicine, Microscope Facility, Department of Medicine, USA
| | - Huanle Yang
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, USA
| | - Jeremy Walston
- Johns Hopkins University, Division of Geriatrics Medicine and Gerontology, USA
| |
Collapse
|
19
|
Bernardi P, Rasola A, Forte M, Lippe G. The Mitochondrial Permeability Transition Pore: Channel Formation by F-ATP Synthase, Integration in Signal Transduction, and Role in Pathophysiology. Physiol Rev 2015; 95:1111-55. [PMID: 26269524 DOI: 10.1152/physrev.00001.2015] [Citation(s) in RCA: 439] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mitochondrial permeability transition (PT) is a permeability increase of the inner mitochondrial membrane mediated by a channel, the permeability transition pore (PTP). After a brief historical introduction, we cover the key regulatory features of the PTP and provide a critical assessment of putative protein components that have been tested by genetic analysis. The discovery that under conditions of oxidative stress the F-ATP synthases of mammals, yeast, and Drosophila can be turned into Ca(2+)-dependent channels, whose electrophysiological properties match those of the corresponding PTPs, opens new perspectives to the field. We discuss structural and functional features of F-ATP synthases that may provide clues to its transition from an energy-conserving into an energy-dissipating device as well as recent advances on signal transduction to the PTP and on its role in cellular pathophysiology.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Andrea Rasola
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Michael Forte
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Giovanna Lippe
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| |
Collapse
|
20
|
Soleimanpour SA, Ferrari AM, Raum JC, Groff DN, Yang J, Kaufman BA, Stoffers DA. Diabetes Susceptibility Genes Pdx1 and Clec16a Function in a Pathway Regulating Mitophagy in β-Cells. Diabetes 2015; 64:3475-84. [PMID: 26085571 PMCID: PMC4587637 DOI: 10.2337/db15-0376] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/02/2015] [Indexed: 12/18/2022]
Abstract
Mitophagy is a critical regulator of mitochondrial quality control and is necessary for elimination of dysfunctional mitochondria to maintain cellular respiration. Here, we report that the homeodomain transcription factor Pdx1, a gene associated with both type 2 diabetes and monogenic diabetes of the young, regulates mitophagy in pancreatic β-cells. Loss of Pdx1 leads to abnormal mitochondrial morphology and function as well as impaired mitochondrial turnover. High-throughput expression microarray and chromatin occupancy analyses reveal that Pdx1 regulates the expression of Clec16a, a type 1 diabetes gene and itself a key mediator of mitophagy through regulation of the E3 ubiquitin ligase Nrdp1. Indeed, expression of Clec16a and Nrdp1 are both reduced in Pdx1 haploinsufficient islets, and reduction of Pdx1 impairs fusion of autophagosomes containing mitochondria to lysosomes during mitophagy. Importantly, restoration of Clec16a expression after Pdx1 loss of function restores mitochondrial trafficking during mitophagy and improves mitochondrial respiration and glucose-stimulated insulin release. Thus, Pdx1 orchestrates nuclear control of mitochondrial function in part by controlling mitophagy through Clec16a. The novel Pdx1-Clec16a-Nrdp1 pathway we describe provides a genetic basis for the pathogenesis of mitochondrial dysfunction in multiple forms of diabetes that could be targeted for future therapies to improve β-cell function.
Collapse
Affiliation(s)
- Scott A Soleimanpour
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Alana M Ferrari
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jeffrey C Raum
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - David N Groff
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Juxiang Yang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Brett A Kaufman
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Doris A Stoffers
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Institute for Diabetes, Obesity and Metabolism of the University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
21
|
Ney PA. Mitochondrial autophagy: Origins, significance, and role of BNIP3 and NIX. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2775-83. [PMID: 25753537 DOI: 10.1016/j.bbamcr.2015.02.022] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 01/24/2015] [Accepted: 02/26/2015] [Indexed: 12/24/2022]
Abstract
Mitochondrial autophagy (mitophagy) is a core cellular activity. In this review, we consider mitophagy and related cellular processes and discuss their significance for human disease. Strong parallels exist between mitophagy and xenophagy employed in host defense. These mechanisms converge on receptors in the innate immune system in clinically relevant scenarios. Mitophagy is part of a cellular quality control mechanism, which is implicated in degenerative disease, especially neurodegenerative disease. Furthermore, mitophagy is an aspect of cellular remodeling, which is employed during development. BNIP3 and NIX are related multi-functional outer mitochondrial membrane proteins. BNIP3 regulates mitophagy during hypoxia, whereas NIX is required for mitophagy during development of the erythroid lineage. Recent advances in the field of BNIP3- and NIX-mediated mitophagy are discussed.
Collapse
Affiliation(s)
- Paul A Ney
- Department of Cell & Molecular Biology, Lindsley F. Kimball Research Institute, New York Blood Center, 310 East 67 Street, New York, NY 10065-6275, USA.
| |
Collapse
|
22
|
Kaufman BA, Li C, Soleimanpour SA. Mitochondrial regulation of β-cell function: maintaining the momentum for insulin release. Mol Aspects Med 2015; 42:91-104. [PMID: 25659350 PMCID: PMC4404204 DOI: 10.1016/j.mam.2015.01.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/29/2015] [Accepted: 01/29/2015] [Indexed: 01/15/2023]
Abstract
All forms of diabetes share the common etiology of insufficient pancreatic β-cell function to meet peripheral insulin demand. In pancreatic β-cells, mitochondria serve to integrate the metabolism of exogenous nutrients into energy output, which ultimately leads to insulin release. As such, mitochondrial dysfunction underlies β-cell failure and the development of diabetes. Mitochondrial regulation of β-cell function occurs through many diverse pathways, including metabolic coupling, generation of reactive oxygen species, maintenance of mitochondrial mass, and through interaction with other cellular organelles. In this chapter, we will focus on the importance of enzymatic regulators of mitochondrial fuel metabolism and control of mitochondrial mass to pancreatic β-cell function, describing how defects in these pathways ultimately lead to diabetes. Furthermore, we will examine the factors responsible for mitochondrial biogenesis and degradation and their roles in the balance of mitochondrial mass in β-cells. Clarifying the causes of β-cell mitochondrial dysfunction may inform new approaches to treat the underlying etiologies of diabetes.
Collapse
Affiliation(s)
- Brett A Kaufman
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Changhong Li
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott A Soleimanpour
- Division of Metabolism, Endocrinology & Diabetes and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Moderate hypoxia induces β-cell dysfunction with HIF-1-independent gene expression changes. PLoS One 2014; 9:e114868. [PMID: 25503986 PMCID: PMC4264765 DOI: 10.1371/journal.pone.0114868] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
Pancreatic β-cell failure is central to the development and progression of type 2 diabetes. We recently demonstrated that β-cells become hypoxic under high glucose conditions due to increased oxygen consumption and that the pancreatic islets of diabetic mice but not those of control mice are moderately hypoxic. However, the impact of moderate hypoxia on β-cell number and function is unknown. In the present study, moderate hypoxia induced a hypoxic response in MIN6 cells, as evidenced by increased levels of HIF-1α protein and target genes. Under these conditions, a selective downregulation of Mafa, Pdx1, Slc2a2, Ndufa5, Kcnj11, Ins1, Wfs1, Foxa2, and Neurod1, which play important roles in β-cells, was also observed in both MIN6 cells and isolated pancreatic islets. Consistent with the altered expression of these genes, abnormal insulin secretion was detected in hypoxic MIN6 cells. Most of the hypoxia-induced gene downregulation in MIN6 cells was not affected by the suppression of HIF-1α, suggesting a HIF-1–independent mechanism. Moderate hypoxia also induced apoptosis in MIN6 cells. These results suggest that hypoxia is a novel stressor of β-cells and that hypoxic stress may play a role in the deterioration of β-cell function.
Collapse
|
24
|
Abstract
Irs2-deficient mice develop type 2-like diabetes due to a reduction in β-cell mass and a failure of pancreatic islets to undergo compensatory hyperplasia in response to insulin resistance. In order to define the molecular mechanisms, we knocked down Irs2 gene expression in mouse MIN6 insulinoma cells. Insulin receptor substrate 2 (IRS2) suppression induced apoptotic cell death, which was associated with an increase in expression of the BH3-only molecule Bim. Knockdown (KD) of Bim reduced apoptotic β-cell death induced by IRS2 suppression. In Irs2-deficient mice, Bim ablation restored β-cell mass, decreased the number of TUNEL-positive cells, and restored normal glucose tolerance after glucose challenge. FoxO1 mediates Bim upregulation induced by IRS2 suppression, and FoxO1 KD partially inhibits β-cell death induced by IRS2 suppression. These results suggest that Bim plays an important role in mediating the increase in β-cell apoptosis and the reduction in β-cell mass that occurs in IRS2-deficient diabetes.
Collapse
Affiliation(s)
- Decheng Ren
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Juan Sun
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Liqun Mao
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Honggang Ye
- Department of Medicine, The University of Chicago, Chicago, IL
| | | |
Collapse
|
25
|
Ren D, Sun J, Wang C, Ye H, Mao L, Cheng EH, Bell GI, Polonsky KS. Role of BH3-only molecules Bim and Puma in β-cell death in Pdx1 deficiency. Diabetes 2014; 63:2744-50. [PMID: 24658302 PMCID: PMC4113059 DOI: 10.2337/db13-1513] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in pancreatic duodenal homeobox-1 (PDX1) are associated with diabetes in humans. Pdx1-haploinsufficient mice develop diabetes due to an increase in β-cell death leading to reduced β-cell mass. For definition of the molecular link between Pdx1 deficiency and β-cell death, Pdx1-haploinsufficient mice in which the genes for the BH3-only molecules Bim and Puma had been ablated were studied on a high-fat diet. Compared with Pdx1(+/-) mice, animals haploinsufficient for both Pdx1 and Bim or Puma genes showed improved glucose tolerance, enhanced β-cell mass, and reduction in the number of TUNEL-positive cells in islets. These results suggest that Bim and Puma ablation improves β-cell survival in Pdx1(+/-) mice. For exploration of the mechanisms responsible for these findings, Pdx1 gene expression was knocked down in mouse MIN6 insulinoma cells resulting in apoptotic cell death that was found to be associated with increased expression of BH3-only molecules Bim and Puma. If the upregulation of Bim and Puma that occurs during Pdx1 suppression was prevented, apoptotic β-cell death was reduced in vitro. These results suggest that Bim and Puma play an important role in β-cell apoptosis in Pdx1-deficient diabetes.
Collapse
Affiliation(s)
- Decheng Ren
- Department of Medicine, University of Chicago, Chicago, IL
| | - Juan Sun
- Department of Medicine, University of Chicago, Chicago, IL
| | | | - Honggang Ye
- Department of Medicine, University of Chicago, Chicago, IL
| | - Liqun Mao
- Department of Medicine, University of Chicago, Chicago, IL
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program and Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Graeme I Bell
- Department of Medicine, University of Chicago, Chicago, IL
| | | |
Collapse
|
26
|
Krautkramer KA, Linnemann AK, Fontaine DA, Whillock AL, Harris TW, Schleis GJ, Truchan NA, Marty-Santos L, Lavine JA, Cleaver O, Kimple ME, Davis DB. Tcf19 is a novel islet factor necessary for proliferation and survival in the INS-1 β-cell line. Am J Physiol Endocrinol Metab 2013; 305:E600-10. [PMID: 23860123 PMCID: PMC3761170 DOI: 10.1152/ajpendo.00147.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recently, a novel type 1 diabetes association locus was identified at human chromosome 6p31.3, and transcription factor 19 (TCF19) is a likely causal gene. Little is known about Tcf19, and we now show that it plays a role in both proliferation and apoptosis in insulinoma cells. Tcf19 is expressed in mouse and human islets, with increasing mRNA expression in nondiabetic obesity. The expression of Tcf19 is correlated with β-cell mass expansion, suggesting that it may be a transcriptional regulator of β-cell mass. Increasing proliferation and decreasing apoptotic cell death are two strategies to increase pancreatic β-cell mass and prevent or delay diabetes. siRNA-mediated knockdown of Tcf19 in the INS-1 insulinoma cell line, a β-cell model, results in a decrease in proliferation and an increase in apoptosis. There was a significant reduction in the expression of numerous cell cycle genes from the late G1 phase through the M phase, and cells were arrested at the G1/S checkpoint. We also observed increased apoptosis and susceptibility to endoplasmic reticulum (ER) stress after Tcf19 knockdown. There was a reduction in expression of genes important for the maintenance of ER homeostasis (Bip, p58(IPK), Edem1, and calreticulin) and an increase in proapoptotic genes (Bim, Bid, Nix, Gadd34, and Pdia2). Therefore, Tcf19 is necessary for both proliferation and survival and is a novel regulator of these pathways.
Collapse
Affiliation(s)
- Kimberly A Krautkramer
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin, Madison, Wisconsin
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yang YHC, Johnson JD. Multi-parameter single-cell kinetic analysis reveals multiple modes of cell death in primary pancreatic β-cells. J Cell Sci 2013; 126:4286-95. [PMID: 23843629 DOI: 10.1242/jcs.133017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Programmed β-cell death plays an important role in both type 1 and type 2 diabetes. Most of what is known about the mechanisms of β-cell death comes from single time-point, single parameter measurements of bulk populations of mixed cells. Such approaches are inadequate for determining the true extent of the heterogeneity in death mechanisms. Here, we characterized the timing and order of molecular events associated with cell death in single β-cells under multiple diabetic stress conditions, including hyperglycemia, cytokine exposure, nutrient deprivation and endoplasmic reticulum (ER) stress. We simultaneously measured the kinetics of six distinct cell death mechanisms by using a caspase-3 sensor and three vital dyes, together with brightfield imaging. We identified several cell death modes where the order of events that usually define apoptosis were not observed. This we termed 'partial apoptosis'. Remarkably, complete classical apoptosis, defined as cells with plasma membrane blebbing, caspase-3 activity, nuclear condensation and membrane annexin V labeling prior to loss of plasma membrane integrity, was found in only half of the cytokine-treated primary β-cells and never in cells stressed by serum removal. By contrast, in the MIN6 cell line, death occurred almost exclusively through complete classical apoptosis. Ambient glucose modulated the cell death mode and kinetics in primary β-cells. Taken together, our data define the kinetic progression of β-cell death mechanisms under different conditions and illustrate the heterogeneity and plasticity of cell death modes in β-cells. We conclude that apoptosis is not the primary mode of adult primary β-cell death.
Collapse
Affiliation(s)
- Yu Hsuan Carol Yang
- Department of Cellular and Physiological Sciences, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver BC V6T 1Z3, Canada
| | | |
Collapse
|
28
|
Chen Y, Decker KF, Zheng D, Matkovich SJ, Jia L, Dorn GW. A nucleus-targeted alternately spliced Nix/Bnip3L protein isoform modifies nuclear factor κB (NFκB)-mediated cardiac transcription. J Biol Chem 2013; 288:15455-65. [PMID: 23603904 DOI: 10.1074/jbc.m113.452342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Several Bcl2 family proteins are expressed both as mitochondrial-targeted full-length and as cytosolic truncated alternately spliced isoforms. Recombinantly expressed shorter Bcl2 family isoforms can heterotypically bind to and prevent mitochondrial localization of their full-length analogs, thus suppressing their activity by sequestration. This "sponge" role requires 1:1 expression stoichiometry; absent this an alternate role is suggested. Here, RNA sequencing revealed coordinate regulation of BH3-only protein Nix/Bnip3L (Nix) and its alternately spliced soluble form (sNix) in hearts, but relative sNix/Nix expression of ∼1:10. Accordingly, we examined other putative functions of sNix. Although Nix expressed in H9c2 rat myoblasts localized to mitochondria, sNix showed variable cytoplasmic and nuclear distribution. Tumor necrosis factor α (TNFα) induced rapid and complete sNix nucleoplasmic translocation concomitant with nuclear translocation of the p65/RelA subunit of NFκB. sNix co-localized and co-precipitated with p65/RelA after TNFα stimulation; TNFα-induced sNix nuclear translocation did not occur in p65/RelA null murine embryonic fibroblasts. ChIP sequencing of TNFα-stimulated H9c2 cells revealed sNix suppression of p65/RelA binding to a subset of weaker DNA binding sites, accounting for its ability to alter gene expression in cultured cells and in vivo mouse hearts. These findings reveal TNFα-stimulated cytoplasmic-nuclear shuttling of the alternately spliced non-mitochondrial Nix isoform and uncover a role for sNix as a modulator of TNFα/NFκB-stimulated cardiac gene expression. Transcriptional co-regulation of sNix and Nix, combined with sNix posttranslational regulation by TNFα, comprises a previously unknown mechanism for molecular cross-talk between extrinsic death receptor and intrinsic mitochondrial apoptosis pathways.
Collapse
Affiliation(s)
- Yun Chen
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Two genes responsible for the juvenile Parkinson’s disease (PD), PINK1 and Parkin, have been implicated in mitochondrial quality control. The inactivation of PINK1, which encodes a mitochondrial kinase, leads to age-dependent mitochondrial degeneration in Drosophila. The phenotype is closely associated with the impairment of mitochondrial respiratory chain activity and defects in mitochondrial dynamics. Drosophila genetic studies have further revealed that PINK1 is an upstream regulator of Parkin and is involved in the mitochondrial dynamics and motility. A series of cell biological studies have given rise to a model in which the activation of PINK1 in damaged mitochondria induces the selective elimination of mitochondria in cooperation with Parkin through the ubiquitin-proteasome and autophagy machineries. Although the relevance of this pathway to PD etiology is still unclear, approaches using stem cells from patients and animal models will help to understand the significance of mitochondrial quality control by the PINK1-Parkin pathway in PD and in healthy individuals. Here I will review recent advances in our understanding of the PINK1-Parkin signaling and will discuss the roles of PINK1-Parkin signaling for mitochondrial maintenance and how the failure of this signaling leads to neurodegeneration.
Collapse
Affiliation(s)
- Yuzuru Imai
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
30
|
Dorn GW. Molecular Mechanisms That Differentiate Apoptosis from Programmed Necrosis. Toxicol Pathol 2012; 41:227-34. [DOI: 10.1177/0192623312466961] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Programmed cell death is physiological when disposing of senescent, dysfunctional, or redundant cells, but pathological if these cells cannot be replaced. Mitochondria help determine cell fate as “gatekeepers” of apoptosis and effectors of cell necrosis. Apoptosis was first described 40 years ago this year. Cell suicide (or the less emotionally charged “programmed cell death”) impacts organism development, normal organ homeostasis, and degenerative (too much cell death) or metaplastic (too little cell death) diseases. The components of apoptosis signaling through mitochondrial targeted Bcl-2 family proteins and activation of the caspase cascade and its downstream proteases and nucleases are well described. More recently, we have realized that there is a parallel cell death pathway, programmed necrosis, in which calcium cross-talk between endoplasmic reticulum and mitochondria causes mitochondrial depolarization, reversal of electron flow through the electron transport chain, and ATP depletion. Since apoptosis and programmed necrosis signaling can occur concurrently in a suicidal cell and are difficult to distinguish using conventional techniques, their relative roles in disease are still being researched and debated. Here, the different molecular mechanisms, effects, and pathophysiological implications of apoptosis and programmed necrosis are reviewed as they relate to heart failure and diabetes mediated by the Bcl-2 family protein, Nix.
Collapse
Affiliation(s)
- Gerald W. Dorn
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
31
|
Sirtuin-4 modulates sensitivity to induction of the mitochondrial permeability transition pore. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1827:38-49. [PMID: 23044393 DOI: 10.1016/j.bbabio.2012.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 09/14/2012] [Accepted: 09/23/2012] [Indexed: 11/20/2022]
Abstract
The sustained opening of the mitochondrial permeability transition pore (PTP) is a decisive event in the onset of irreversible cell injury. The PTP is modulated by numerous exogenous and endogenous effectors, including mitochondrial membrane potential, ions and metabolites. Mitochondrial sirtuins have recently emerged as pivotal mediators of mitochondrial metabolism. In the present study, we demonstrate that sirt-4 modulates sensitivity to PTP onset induced by calcium and the oxidative cross linking reagent phenylarsine oxide, and PTP dependent cytotoxicity brought about by TNF or doxorubicin. Moreover, the ability of sirt-4 to modulate onset of the PTP is dependent on the expression of glutamate dehydrogenase-1.
Collapse
|
32
|
Inflammation-Mediated Regulation of MicroRNA Expression in Transplanted Pancreatic Islets. J Transplant 2012; 2012:723614. [PMID: 22655170 PMCID: PMC3359768 DOI: 10.1155/2012/723614] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/09/2012] [Accepted: 02/20/2012] [Indexed: 12/22/2022] Open
Abstract
Nonspecific inflammation in the transplant microenvironment results in β-cell dysfunction and death influencing negatively graft outcome. MicroRNA (miRNA) expression and gene target regulation in transplanted islets are not yet well characterized. We evaluated the impact of inflammation on miRNA expression in transplanted rat islets. Islets exposed in vitro to proinflammatory cytokines and explanted syngeneic islet grafts were evaluated by miRNA arrays. A subset of 26 islet miRNAs was affected by inflammation both in vivo and in vitro. Induction of miRNAs was dependent on NF-κB, a pathway linked with cytokine-mediated islet cell death. RT-PCR confirmed expression of 8 miRNAs. The association between these miRNAs and mRNA target-predicting algorithms in genome-wide RNA studies of β-cell inflammation identified 238 potential miRNA gene targets. Several genes were ontologically associated with regulation of insulin signaling and secretion, diabetes, and islet physiology. One of the most activated miRNAs was miR-21. Overexpression of miR-21 in insulin-secreting MIN6 cells downregulated endogenous expression of the tumor suppressor Pdcd4 and of Pclo, a Ca2+ sensor protein involved in insulin secretion. Bioinformatics identified both as potential targets. The integrated analysis of miRNA and mRNA expression profiles revealed potential targets that may identify molecular targets for therapeutic interventions.
Collapse
|
33
|
Bedoya FJ, Salguero-Aranda C, Cahuana GM, Tapia-Limonchi R, Soria B, Tejedo JR. Regulation of pancreatic β-cell survival by nitric oxide: clinical relevance. Islets 2012; 4:108-18. [PMID: 22614339 DOI: 10.4161/isl.19822] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The reduction of pancreatic β-cell mass is an important factor in the development of type 1 and type 2 diabetes. Understanding the mechanisms that regulate the maintenance of pancreatic β-cell mass as well as β-cell death is necessary for the establishment of therapeutic strategies. In this context, nitric oxide (NO) is a diatomic, gaseous, highly reactive molecule with biological activity that participates in the regulation of pancreatic β-cell mass. Two types of cellular responses can be distinguished depending on the level of NO production. First, pancreatic β-cells exposed to inflammatory cytokines, lipid stress or hyperglycaemia produce high concentrations of NO, mainly due to the activation of inducible NO synthase (iNOS), thus promoting cell death. Meanwhile, under homeostatic conditions, low concentrations of NO, constitutively produced by endothelial NO synthase (eNOS), promote cell survival. Here, we will discuss the current knowledge of the NO-dependent mechanisms activated during cellular responses, emphasizing those related to the regulation of cell survival.
Collapse
Affiliation(s)
- Francisco J Bedoya
- Andalusian Center for Molecular Biology and Regenerative Medicine, University Pablo de Olavide, CIBERDEM, RED-TERCEL, Seville, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Khoo C, Yang J, Weinrott SA, Kaestner KH, Naji A, Schug J, Stoffers DA. Research resource: the pdx1 cistrome of pancreatic islets. Mol Endocrinol 2012; 26:521-33. [PMID: 22322596 DOI: 10.1210/me.2011-1231] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The homeodomain transcription factor pancreas duodenal homeobox 1 (Pdx1, also known as insulin promoter factor 1) is a master regulator of pancreas development, as mice or humans lacking Pdx1 function are a pancreatic. Importantly, heterozygous mutations in Pdx1 cause early and late onset forms of diabetes in humans. Despite these central roles in development and adult β-cell function, we have only rudimentary knowledge of the transcriptome targets of Pdx1 that mediate these phenotypes. Therefore, we performed global location analysis of Pdx1 occupancy in pancreatic islets. We used evolutionary conservation of target genes to identify the most relevant Pdx1 targets by performing chromatin immunoprecipitation sequencing on both human and mouse islets. Remarkably, the conserved target set is highly enriched for genes annotated to function in endocrine system and metabolic disorders, various signaling pathways, and cell survival, providing a molecular explanation for many of the phenotypes resulting from Pdx1 deficiency.
Collapse
Affiliation(s)
- Cynthia Khoo
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Akirav EM, Lebastchi J, Galvan EM, Henegariu O, Akirav M, Ablamunits V, Lizardi PM, Herold KC. Detection of β cell death in diabetes using differentially methylated circulating DNA. Proc Natl Acad Sci U S A 2011; 108:19018-23. [PMID: 22074781 PMCID: PMC3223447 DOI: 10.1073/pnas.1111008108] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In diabetes mellitus, β cell destruction is largely silent and can be detected only after significant loss of insulin secretion capacity. We have developed a method for detecting β cell death in vivo by amplifying and measuring the proportion of insulin 1 DNA from β cells in the serum. By using primers that are specific for DNA methylation patterns in β cells, we have detected circulating copies of β cell-derived demethylated DNA in serum of mice by quantitative PCR. Accordingly, we have identified a relative increase of β cell-derived DNA after induction of diabetes with streptozotocin and during development of diabetes in nonobese diabetic mice. We have extended the use of this assay to measure β cell-derived insulin DNA in human tissues and serum. We found increased levels of demethylated insulin DNA in subjects with new-onset type 1 diabetes compared with age-matched control subjects. Our method provides a noninvasive approach for detecting β cell death in vivo that may be used to track the progression of diabetes and guide its treatment.
Collapse
Affiliation(s)
- Eitan M. Akirav
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| | - Jasmin Lebastchi
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| | - Eva M. Galvan
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| | - Octavian Henegariu
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| | - Michael Akirav
- Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel; and
| | - Vitaly Ablamunits
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| | - Paul M. Lizardi
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06511
| | - Kevan C. Herold
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06511
| |
Collapse
|
36
|
Affiliation(s)
- Stefan S Fajans
- Department of Internal Medicine, Universityof Michigan Health System, Ann Arbor, MI, USA.
| | | |
Collapse
|