1
|
Rajkumari N, Shalayel I, Tubbs E, Perrier Q, Chabert C, Lablanche S, Benhamou PY, Arnol C, Gredy L, Divoux T, Stephan O, Zebda A, van der Sanden B. Matrix design for optimal pancreatic β cells transplantation. BIOMATERIALS ADVANCES 2024; 164:213980. [PMID: 39126900 DOI: 10.1016/j.bioadv.2024.213980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024]
Abstract
New therapeutic approaches to treat type 1 diabetes mellitus relies on pancreatic islet transplantation. Here, developing immuno-isolation strategies is essential to eliminate the need for systemic immunosuppression after pancreatic islet grafts. A solution is the macro-encapsulation of grafts in semipermeable matrixes with a double function: separating islets from host immune cells and facilitating the diffusion of insulin, glucose, and other metabolites. This study aims to synthesize and characterize different types of gelatin-collagen matrixes to prepare a macro-encapsulation device for pancreatic islets that fulfill these functions. While natural polymers exhibit superior biocompatibility compared to synthetic ones, their mechanical properties are challenging to reproduce. To address this issue, we conducted a comparative analysis between photo-crosslinked gelatin matrixes and chemically crosslinked collagen matrixes. We show that the different crosslinkers and polymerization methods influence the survival and glucose-stimulated insulin production of pancreatic β cells (INS1) in vitro, as well as the in vitro and in vivo stability of the matrix and the immuno-isolation in vivo. Among the matrixes, the stiff multilayer GelMA matrixes (8.5 kPa), fabricated by digital light processing, were the best suited for pancreatic β cells macro-encapsulation regarding these parameters. Within the alveoli of this matrix, pancreatic β cells spontaneously formed aggregates.
Collapse
Affiliation(s)
- Nikita Rajkumari
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble Alpes University and INSERM U1055, France; Nantes University, CRCI2NA, INSERM 1307, 44000 Nantes, France.
| | - Ibrahim Shalayel
- SyNaBi & Platform of Intravital Microscopy, TIMC-IMAG, CNRS UMR 5525, Grenoble Alpes University, Grenoble INP, INSERM, Grenoble, France.
| | - Emily Tubbs
- Grenoble Alpes University, CEA, INSERM, IRIG, 38000 Grenoble, Biomics, France.
| | - Quentin Perrier
- Univ. Grenoble Alpes, INSERM, Grenoble Alpes University Hospital, Department of Pharmacy, LBFA U1055, Grenoble, France.
| | - Clovis Chabert
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble Alpes University and INSERM U1055, France.
| | - Sandrine Lablanche
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble Alpes University and INSERM U1055, France; Department of Endocrinology-Diabetology-Nutrition, Grenoble University Hospital, France.
| | - Pierre-Yves Benhamou
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble Alpes University and INSERM U1055, France; Department of Endocrinology-Diabetology-Nutrition, Grenoble University Hospital, France.
| | - Capucine Arnol
- SyNaBi & Platform of Intravital Microscopy, TIMC-IMAG, CNRS UMR 5525, Grenoble Alpes University, Grenoble INP, INSERM, Grenoble, France
| | - Laetitia Gredy
- MoVe, Laboratoire interdisciplinaire de physique, CNRS UMR 5588, Grenoble Alpes University, St-Martin d'Hères, France.
| | - Thibaut Divoux
- ENSL, CNRS, Laboratoire de Physique, F-69342 Lyon, France.
| | - Olivier Stephan
- MoVe, Laboratoire interdisciplinaire de physique, CNRS UMR 5588, Grenoble Alpes University, St-Martin d'Hères, France.
| | - Abdelkader Zebda
- SyNaBi & Platform of Intravital Microscopy, TIMC-IMAG, CNRS UMR 5525, Grenoble Alpes University, Grenoble INP, INSERM, Grenoble, France.
| | - Boudewijn van der Sanden
- SyNaBi & Platform of Intravital Microscopy, TIMC-IMAG, CNRS UMR 5525, Grenoble Alpes University, Grenoble INP, INSERM, Grenoble, France.
| |
Collapse
|
2
|
Reed J, Higginbotham V, Bain S, Kanamarlapudi V. Comparative Analysis of Orthosteric and Allosteric GLP-1R Agonists' Effects on Insulin Secretion from Healthy, Diabetic, and Recovered INS-1E Pancreatic Beta Cells. Int J Mol Sci 2024; 25:6331. [PMID: 38928038 PMCID: PMC11203424 DOI: 10.3390/ijms25126331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Despite the availability of different treatments for type 2 diabetes (T2D), post-diagnosis complications remain prevalent; therefore, more effective treatments are desired. Glucagon-like peptide (GLP)-1-based drugs are currently used for T2D treatment. They act as orthosteric agonists for the GLP-1 receptor (GLP-1R). In this study, we analyzed in vitro how the GLP-1R orthosteric and allosteric agonists augment glucose-stimulated insulin secretion (GSIS) and intracellular cAMP production (GSICP) in INS-1E pancreatic beta cells under healthy, diabetic, and recovered states. The findings from this study suggest that allosteric agonists have a longer duration of action than orthosteric agonists. They also suggest that the GLP-1R agonists do not deplete intracellular insulin, indicating they can be a sustainable and safe treatment option for T2D. Importantly, this study demonstrates that the GLP-1R agonists variably augment GSIS through GSICP in healthy, diabetic, and recovered INS-1E cells. Furthermore, we find that INS-1E cells respond differentially to the GLP-1R agonists depending on both glucose concentration during and before treatment and/or whether the cells have been previously exposed to these drugs. In conclusion, the findings described in this manuscript will be useful in determining in vitro how pancreatic beta cells respond to T2D drug treatments in healthy, diabetic, and recovered states.
Collapse
Affiliation(s)
| | | | | | - Venkateswarlu Kanamarlapudi
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK; (J.R.); (V.H.); (S.B.)
| |
Collapse
|
3
|
Sakayanathan P, Loganathan C, Thayumanavan P. Protection of pancreatic beta cells against high glucose-induced toxicity by astaxanthin-s-allyl cysteine diester: alteration of oxidative stress and apoptotic-related protein expression. Arch Physiol Biochem 2024; 130:316-324. [PMID: 35482540 DOI: 10.1080/13813455.2022.2064878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/18/2022] [Accepted: 04/05/2022] [Indexed: 11/02/2022]
Abstract
Purpose: High glucose (HG)-induced oxidative stress is associated with apoptosis in pancreatic β-cells. The protective effect of astaxanthin-s-allyl cysteine diester (AST-SAC) against HG-induced oxidative stress in pancreatic β-cells (βTC-tet cell line) in in vitro was studied.Materials and Methods: βTC-tet cell line was exposed to HG in the presence and absence of AST-SAC. Various parameters such as cell viability, reactive oxygen species generation, mitochondrial membrane potential, DNA fragmentation and expression of proteins involved in apoptosis [p53, B-cell lymphoma 2 (Bcl-2), Bcl-2 associated X (Bax), cytochrome c and caspase 3] were studied.Results: Pre-treatment of βTC-tet cells with AST-SAC (4, 8 and 12 μg/ml) in the presence of HG (25 mM) protected the viability of the cells in a dose-dependent manner. AST-SAC treatment mitigated the oxidative stress thereby preventing the mitochondrial dysfunction, DNA damage and apoptosis in βTC-tet cells against HG toxicity. Treatment with AST-SAC prevented the increased expression of p53 under HG conditions. Further, AST-SAC treatment maintained the level of pro-apoptotic (Bax, cleaved caspase-3 and cytochrome c) and anti-apoptotic (Bcl-2) proteins to that of the control level under HG exposed conditions in βTC-tet cells.Conclusion: Altogether, AST-SAC alleviated HG-induced oxidative damage and apoptosis in pancreatic β-cells by enhancing the antioxidant status and altering apoptotic-related protein expression.
Collapse
Affiliation(s)
| | - Chitra Loganathan
- Department of Biochemistry, Periyar University, Salem, India
- Research and Development center, Bioinnov Solutions LLP, Salem, India
| | | |
Collapse
|
4
|
Li Y, Lou N, Liu X, Zhuang X, Chen S. Exploring new mechanisms of Imeglimin in diabetes treatment: Amelioration of mitochondrial dysfunction. Biomed Pharmacother 2024; 175:116755. [PMID: 38772155 DOI: 10.1016/j.biopha.2024.116755] [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: 12/17/2023] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
With the increasing prevalence of type 2 diabetes mellitus (T2DM), it has become critical to identify effective treatment strategies. In recent years, the novel oral hypoglycaemic drug Imeglimin has attracted much attention in the field of diabetes treatment. The mechanisms of its therapeutic action are complex and are not yet fully understood by current research. Current evidence suggests that pancreatic β-cells, liver, and skeletal muscle are the main organs in which Imeglimin lowers blood glucose levels and that it acts mainly by targeting mitochondrial function, thereby inhibiting hepatic gluconeogenesis, enhancing insulin sensitivity, promoting pancreatic β-cell function, and regulating energy metabolism. There is growing evidence that the drug also has a potentially volatile role in the treatment of diabetic complications, including metabolic cardiomyopathy, diabetic vasculopathy, and diabetic neuroinflammation. According to available clinical studies, its efficacy and safety profile are more evident than other hypoglycaemic agents, and it has synergistic effects when combined with other antidiabetic drugs, and also has potential in the treatment of T2DM-related complications. This review aims to shed light on the latest research progress in the treatment of T2DM with Imeglimin, thereby providing clinicians and researchers with the latest insights into Imeglimin as a viable option for the treatment of T2DM.
Collapse
Affiliation(s)
- Yilin Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Nenngjun Lou
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xiaojing Liu
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xianghua Zhuang
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| | - Shihong Chen
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| |
Collapse
|
5
|
Ngamjariyawat A, Cen J, Wang X, Welsh N. GDF15 Protects Insulin-Producing Beta Cells against Pro-Inflammatory Cytokines and Metabolic Stress via Increased Deamination of Intracellular Adenosine. Int J Mol Sci 2024; 25:801. [PMID: 38255875 PMCID: PMC10815691 DOI: 10.3390/ijms25020801] [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: 12/04/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
It has been proposed that antidiabetic drugs, such as metformin and imatinib, at least in part, promote improved glucose tolerance in type 2 diabetic patients via increased production of the inflammatory cytokine GDF15. This is supported by studies, performed in rodent cell lines and mouse models, in which the addition or production of GDF15 improved beta-cell function and survival. The aim of the present study was to determine whether human beta cells produce GDF15 in response to antidiabetic drugs and, if so, to further elucidate the mechanisms by which GDF15 modulates the function and survival of such cells. The effects and expression of GDF15 were analyzed in human insulin-producing EndoC-betaH1 cells and human islets. We observed that alpha and beta cells exhibit considerable heterogeneity in GDF15 immuno-positivity. The predominant form of GDF15 present in islet and EndoC-betaH1 cells was pro-GDF15. Imatinib, but not metformin, increased pro-GDF15 levels in EndoC-betaH1 cells. Under basal conditions, exogenous GDF15 increased human islet oxygen consumption rates. In EndoC-betaH1 cells and human islets, exogenous GDF15 partially ameliorated cytokine- or palmitate + high-glucose-induced loss of function and viability. GDF15-induced cell survival was paralleled by increased inosine levels, suggesting a more efficient disposal of intracellular adenosine. Knockdown of adenosine deaminase, the enzyme that converts adenosine to inosine, resulted in lowered inosine levels and loss of protection against cytokine- or palmitate + high-glucose-induced cell death. It is concluded that imatinib-induced GDF15 production may protect human beta cells partially against inflammatory and metabolic stress. Furthermore, it is possible that the GDF15-mediated activation of adenosine deaminase and the increased disposal of intracellular adenosine participate in protection against beta-cell death.
Collapse
Affiliation(s)
- Anongnad Ngamjariyawat
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden; (A.N.); (J.C.); (X.W.)
- Division of Anatomy, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Jing Cen
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden; (A.N.); (J.C.); (X.W.)
| | - Xuan Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden; (A.N.); (J.C.); (X.W.)
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden; (A.N.); (J.C.); (X.W.)
| |
Collapse
|
6
|
Li J, Jiang Q, Wang X, Hou L, Wang L, Lou K, Pang S. Metformin Preserves Insulin Secretion in Pancreatic β-cells through FGF21/Akt Pathway In vitro and In vivo. Comb Chem High Throughput Screen 2024; 27:2691-2698. [PMID: 37855357 DOI: 10.2174/0113862073246747230920170201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/09/2023] [Accepted: 08/06/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND In our previous studies, it was found that metformin can elevate the expression of FGF21 in the peripheral blood of type 2 diabetic rats and improve insulin sensitivity in diabetic rats. However, whether this effect is mediated by increased FGF21 expression in pancreatic islet β-cells is still unknown. Therefore, this study focuses on the effect of metformin on insulin secretion in pancreatic β-cells. AIMS Metformin can effectivly improve insulin resistance. Metformin influencing pancreatic β- cell function is inclusive. In this study, we sought to analyze possible variations in insulin secretion and possible signaling mechanisms after metformin intervention. METHODS The study employed an in vivo model of a high-fat diet in streptozocin-induced diabetic rats and an in vitro model of rat pancreatic β-cells (INS-1 cells) that were subjected to damage caused by hyperglycemia and hyperlipidemia. After treating INS-1 cells in normal, high-glucose, and high-glucose+metformin, we measured insulin secretion by glucose-stimulated insulin secretion (GSIS). Insulin was measured using an enzyme-linked immunosorbent assay. FGF21 expression was detected by RT-PCR and Western blot, as well as that p-Akt and t-Akt expression were detected by Western blot in INS-1 cells and diabetic rat islets. Finally, to verify the regulation of the FGF21 /Akt axis in metformin administration, additional experiments were carried out in metformin-stimulated INS-1 cells. RESULTS High-glucose could significantly stimulate insulin secretion while metformin preserved insulin secretion. Expression of FGF21 and p-Akt was decreased in high-glucose, however, metformin could reverse this effect in INS-1 cells and diabetic rat islets. CONCLUSION Our results demonstrate a protective role of metformin in preserving insulin secretion through FGF21/Akt signaling in T2DM.
Collapse
Affiliation(s)
- Jianting Li
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Qiang Jiang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Xin Wang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Lulu Hou
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Lulu Wang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Kai Lou
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Shuguang Pang
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
- Department of Clinical Medicine, Weifang Medical College, Weifang, 261000, China
| |
Collapse
|
7
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
8
|
Mu-U-Min RBA, Diane A, Allouch A, Al-Siddiqi HH. Ca 2+-Mediated Signaling Pathways: A Promising Target for the Successful Generation of Mature and Functional Stem Cell-Derived Pancreatic Beta Cells In Vitro. Biomedicines 2023; 11:1577. [PMID: 37371672 DOI: 10.3390/biomedicines11061577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes mellitus is a chronic disease affecting over 500 million adults globally and is mainly categorized as type 1 diabetes mellitus (T1DM), where pancreatic beta cells are destroyed, and type 2 diabetes mellitus (T2DM), characterized by beta cell dysfunction. This review highlights the importance of the divalent cation calcium (Ca2+) and its associated signaling pathways in the proper functioning of beta cells and underlines the effects of Ca2+ dysfunction on beta cell function and its implications for the onset of diabetes. Great interest and promise are held by human pluripotent stem cell (hPSC) technology to generate functional pancreatic beta cells from diabetic patient-derived stem cells to replace the dysfunctional cells, thereby compensating for insulin deficiency and reducing the comorbidities of the disease and its associated financial and social burden on the patient and society. Beta-like cells generated by most current differentiation protocols have blunted functionality compared to their adult human counterparts. The Ca2+ dynamics in stem cell-derived beta-like cells and adult beta cells are summarized in this review, revealing the importance of proper Ca2+ homeostasis in beta-cell function. Consequently, the importance of targeting Ca2+ function in differentiation protocols is suggested to improve current strategies to use hPSCs to generate mature and functional beta-like cells with a comparable glucose-stimulated insulin secretion (GSIS) profile to adult beta cells.
Collapse
Affiliation(s)
- Razik Bin Abdul Mu-U-Min
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Asma Allouch
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Heba H Al-Siddiqi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| |
Collapse
|
9
|
Vanillic acid potentiates insulin secretion and prevents pancreatic β-cells cytotoxicity under H 2O 2-induced oxidative stress. Mol Biol Rep 2023; 50:1311-1320. [PMID: 36454432 DOI: 10.1007/s11033-022-08046-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/19/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Oxidative stress is known to impair cellular functions and, therefore, plays a significant role in the pathophysiology of various diseases, including diabetes. The persistently elevated glucose levels may cause enhanced mitochondrial reactive oxygen species generation, which in turn can damage the pancreatic β-cells. In this study, we have investigated the effect of vanillic acid on preventing H2O2-induced β-cells death and retaining its insulin secretion potentiating effect in the presence of H2O2. METHODS The insulin secretion from the BRIN-BD11 cells was quantified using ELISA-based assays. The viability of the cells was assessed by estimated by the [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] (MTT) colorimetric assay and DAPI staining. The expression levels of apoptotic and antioxidant proteins were estimated by western blot experiments. RESULTS Vanillic acid protected pancreatic β-cells viability and function under the H2O2 oxidative stress condition. The Erk1/2 activation appears to play an important role in vanillic acid potentiated insulin secretion and protection of the β-cells in the presence of H2O2. Vanillic acid pretreated cells exhibited enhanced expression of antioxidant enzymes such as catalase and SOD-2 and reduced the expression of proapoptotic markers such as BAX and BAD. In addition, it also enhanced the expression of oxidative stress-sensitive transcription factor Nrf-2 and cell survival protein Akt. CONCLUSION The present study shows that vanillic acid potentiates insulin secretion and protects pancreatic β-cells from H2O2-induced oxidative stress.
Collapse
|
10
|
Starinets VS, Serov DA, Penkov NV, Belosludtseva NV, Dubinin MV, Belosludtsev KN. Alisporivir Normalizes Mitochondrial Function of Primary Mouse Lung Endothelial Cells Under Conditions of Hyperglycemia. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:605-616. [PMID: 36154883 PMCID: PMC9282907 DOI: 10.1134/s0006297922070033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/16/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022]
Abstract
Effect of alisporivir (a mitochondrial permeability transition pore inhibitor) on the development of mitochondrial dysfunction under hyperglycemic conditions in the primary culture of mouse lung endothelial cells was investigated in this work. We demonstrated that hyperglycemia (30 mM glucose for 24 h) leads to the decrease in viability of the pulmonary endotheliocytes, causes mitochondrial dysfunction manifested by the drop in membrane potential and increase in superoxide anion generation as well as facilitates opening of the mitochondrial permeability transition pore (MPT pore). Incubation of endothelial cells with 5 µM alisporivir under hyperglycemic conditions leads to the increase in cell viability, restoration of the membrane potential level and of the MPT pore opening activity to control values. Hyperglycemia causes increased mitophagy in the lung endothelial cells: we observed increase in the degree of colocalization of mitochondria and lysosomes and upregulation of the Parkin gene expression. Alisporivir restores these parameters back to the levels observed in the control cells. Hyperglycemia results in the increase in the expression of the Drp1 gene in endotheliocytes responsible for synthesis of the protein involved in the process of mitochondria fission. Alisporivir does not significantly alter expression of the genes. The paper discusses mechanisms of the effect of alisporivir on mitochondrial dysfunction in murine pulmonary endotheliocytes under conditions of hyperglycemia.
Collapse
Affiliation(s)
- Vlada S Starinets
- Mari State University, Yoshkar-Ola, 424001, Mari El, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Dmitriy A Serov
- Biophotonics Center, Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, 119991, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Nikita V Penkov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Natalia V Belosludtseva
- Mari State University, Yoshkar-Ola, 424001, Mari El, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | | | - Konstantin N Belosludtsev
- Mari State University, Yoshkar-Ola, 424001, Mari El, Russia.
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
11
|
Кузнецов КО, Саетова АА, Махмутова ЭИ, Бобрик АГ, Бобрик ДВ, Нагаев ИР, Хамитова АД, Арапиева АМ. [Imeglimin: features of the mechanism of action and potential benefits]. PROBLEMY ENDOKRINOLOGII 2022; 68:57-66. [PMID: 35841169 PMCID: PMC9762543 DOI: 10.14341/probl12868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/25/2022] [Accepted: 03/11/2022] [Indexed: 06/15/2023]
Abstract
Imeglimin is the first drug in a new class of tetrahydrotriazine-containing oral hypoglycemic agents called «glimines». Its mechanism of action is aimed at achieving a double effect, firstly, to improve the function of beta cells of the pancreas, and secondly, to enhance the action of insulin in key tissues, including the liver and skeletal muscles. At the cellular level, imeglimin modulates mitochondrial function, which leads to an improvement in cellular energy metabolism, as well as to the protection of cells from death in conditions of excessive accumulation of reactive oxygen species. It is important to note that the mechanism of action of imeglimin differs from existing drugs used for the treatment of type 2 diabetes mellitus. Like glucagon-like peptide-1 receptor agonists, imeglimin enhances insulin secretion in an exclusively glucose-dependent manner, but their mechanism of action at the cellular level diverges. Sulfonylureas and glinides function by closing ATP-sensitive potassium channels to release insulin, which is also different from imeglimin. Compared with metformin, the effect of imeglimine is also significantly different. Other major classes of oral antihypertensive agents, such as sodium-glucose transporter-2 inhibitors, thiazolidinediones and α glucosidase inhibitors mediate their action through mechanisms that do not overlap with imeglimine. Given such differences in the mechanisms of action, imeglimin can be used as part of combination therapy, for example with sitagliptin and metformin. The imeglimine molecule is well absorbed (Tmax-4), and the half-life is 5-6 hours, is largely excreted through the kidneys, and also has no clinically significant interactions with either metformin or sitagliptin.
Collapse
Affiliation(s)
- К. О. Кузнецов
- Российский национальный исследовательский медицинский университет им. Н.И. Пирогова
| | | | | | - А. Г. Бобрик
- Башкирский государственный медицинский университет
| | - Д. В. Бобрик
- Башкирский государственный медицинский университет
| | - И. Р. Нагаев
- Башкирский государственный медицинский университет
| | | | | |
Collapse
|
12
|
Newsholme P, Rowlands J, Rose’Meyer R, Cruzat V. Metabolic Adaptions/Reprogramming in Islet Beta-Cells in Response to Physiological Stimulators—What Are the Consequences. Antioxidants (Basel) 2022; 11:antiox11010108. [PMID: 35052612 PMCID: PMC8773416 DOI: 10.3390/antiox11010108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/25/2022] Open
Abstract
Irreversible pancreatic β-cell damage may be a result of chronic exposure to supraphysiological glucose or lipid concentrations or chronic exposure to therapeutic anti-diabetic drugs. The β-cells are able to respond to blood glucose in a narrow concentration range and release insulin in response, following activation of metabolic pathways such as glycolysis and the TCA cycle. The β-cell cannot protect itself from glucose toxicity by blocking glucose uptake, but indeed relies on alternative metabolic protection mechanisms to avoid dysfunction and death. Alteration of normal metabolic pathway function occurs as a counter regulatory response to high nutrient, inflammatory factor, hormone or therapeutic drug concentrations. Metabolic reprogramming is a term widely used to describe a change in regulation of various metabolic enzymes and transporters, usually associated with cell growth and proliferation and may involve reshaping epigenetic responses, in particular the acetylation and methylation of histone proteins and DNA. Other metabolic modifications such as Malonylation, Succinylation, Hydroxybutyrylation, ADP-ribosylation, and Lactylation, may impact regulatory processes, many of which need to be investigated in detail to contribute to current advances in metabolism. By describing multiple mechanisms of metabolic adaption that are available to the β-cell across its lifespan, we hope to identify sites for metabolic reprogramming mechanisms, most of which are incompletely described or understood. Many of these mechanisms are related to prominent antioxidant responses. Here, we have attempted to describe the key β-cell metabolic adaptions and changes which are required for survival and function in various physiological, pathological and pharmacological conditions.
Collapse
Affiliation(s)
- Philip Newsholme
- Curtin Medical School and CHIRI, Curtin University, Perth, WA 6845, Australia
- Correspondence: (P.N.); (J.R.)
| | - Jordan Rowlands
- Curtin Medical School and CHIRI, Curtin University, Perth, WA 6845, Australia
- Correspondence: (P.N.); (J.R.)
| | - Roselyn Rose’Meyer
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Vinicius Cruzat
- Faculty of Health, Torrens University Australia, Brisbane, QLD 4006, Australia;
| |
Collapse
|
13
|
Shoshan-Barmatz V, Anand U, Nahon-Crystal E, Di Carlo M, Shteinfer-Kuzmine A. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target? Front Physiol 2021; 12:730048. [PMID: 34671273 PMCID: PMC8521008 DOI: 10.3389/fphys.2021.730048] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin's effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin's adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - Marta Di Carlo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
14
|
Mitochondrial Calcium Signaling in Pancreatic β-Cell. Int J Mol Sci 2021; 22:ijms22052515. [PMID: 33802289 PMCID: PMC7959128 DOI: 10.3390/ijms22052515] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulation of calcium in energized mitochondria of pancreatic β-cells is emerging as a crucial process for pancreatic β-cell function. β-cell mitochondria sense and shape calcium signals, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion during nutrient stimulation. Here, we describe the role of mitochondrial calcium signaling in pancreatic β-cell function. We report the latest pharmacological and genetic findings, including the first mitochondrial calcium-targeted intervention strategies developed to modulate pancreatic β-cell function and their potential relevance in the context of diabetes.
Collapse
|
15
|
Hallakou‐Bozec S, Vial G, Kergoat M, Fouqueray P, Bolze S, Borel A, Fontaine E, Moller DE. Mechanism of action of Imeglimin: A novel therapeutic agent for type 2 diabetes. Diabetes Obes Metab 2021; 23:664-673. [PMID: 33269554 PMCID: PMC8049051 DOI: 10.1111/dom.14277] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/15/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023]
Abstract
Imeglimin is an investigational first-in-class novel oral agent for the treatment of type 2 diabetes (T2D). Several pivotal phase III trials have been completed with evidence of statistically significant glucose lowering and a generally favourable safety and tolerability profile, including the lack of severe hypoglycaemia. Imeglimin's mechanism of action involves dual effects: (a) amplification of glucose-stimulated insulin secretion (GSIS) and preservation of β-cell mass; and (b) enhanced insulin action, including the potential for inhibition of hepatic glucose output and improvement in insulin signalling in both liver and skeletal muscle. At a cellular and molecular level, Imeglimin's underlying mechanism may involve correction of mitochondrial dysfunction, a common underlying element of T2D pathogenesis. It has been observed to rebalance respiratory chain activity (partial inhibition of Complex I and correction of deficient Complex III activity), resulting in reduced reactive oxygen species formation (decreasing oxidative stress) and prevention of mitochondrial permeability transition pore opening (implicated in preventing cell death). In islets derived from diseased rodents with T2D, Imeglimin also enhances glucose-stimulated ATP generation and induces the synthesis of nicotinamide adenine dinucleotide (NAD+ ) via the 'salvage pathway'. In addition to playing a key role as a mitochondrial co-factor, NAD+ metabolites may contribute to the increase in GSIS (via enhanced Ca++ mobilization). Imeglimin has also been shown to preserve β-cell mass in rodents with T2D. Overall, Imeglimin appears to target a key root cause of T2D: defective cellular energy metabolism. This potential mode of action is unique and has been shown to differ from that of other major therapeutic classes, including biguanides, sulphonylureas and glucagon-like peptide-1 receptor agonists.
Collapse
Affiliation(s)
| | - Guillaume Vial
- Université Grenoble AlpesGrenobleFrance
- Inserm U 1042, Laboratoire INSERM U1042, Hypoxia PathoPhysiology (HP2)GrenobleFrance
| | | | | | | | - Anne‐Laure Borel
- Université Grenoble AlpesGrenobleFrance
- Inserm U 1042, Laboratoire INSERM U1042, Hypoxia PathoPhysiology (HP2)GrenobleFrance
- Centre Hospitalier Universitaire Grenoble Alpes, département de Endocrinologie‐diabétologie‐Nutrition, Centre Spécialisé de l'Obésité Grenoble Arc AlpinGrenobleFrance
| | - Eric Fontaine
- Université Grenoble Alpes, LBFAGrenobleFrance
- Inserm U 1055, LBFAGrenobleFrance
| | | |
Collapse
|
16
|
Georgiadou E, Rutter GA. Control by Ca 2+ of mitochondrial structure and function in pancreatic β-cells. Cell Calcium 2020; 91:102282. [PMID: 32961506 PMCID: PMC7116533 DOI: 10.1016/j.ceca.2020.102282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Mitochondria play a central role in glucose metabolism and the stimulation of insulin secretion from pancreatic β-cells. In this review, we discuss firstly the regulation and roles of mitochondrial Ca2+ transport in glucose-regulated insulin secretion, and the molecular machinery involved. Next, we discuss the evidence that mitochondrial dysfunction in β-cells is associated with type 2 diabetes, from a genetic, functional and structural point of view, and then the possibility that these changes may in part be mediated by dysregulation of cytosolic Ca2+. Finally, we review the importance of preserved mitochondrial structure and dynamics for mitochondrial gene expression and their possible relevance to the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
17
|
Belosludtsev KN, Belosludtseva NV, Dubinin MV. Diabetes Mellitus, Mitochondrial Dysfunction and Ca 2+-Dependent Permeability Transition Pore. Int J Mol Sci 2020; 21:6559. [PMID: 32911736 PMCID: PMC7555889 DOI: 10.3390/ijms21186559] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic diseases in the developed world, and is associated either with the impaired secretion of insulin or with the resistance of cells to the actions of this hormone (type I and type II diabetes, respectively). In both cases, a common pathological change is an increase in blood glucose-hyperglycemia, which eventually can lead to serious damage to the organs and tissues of the organism. Mitochondria are one of the main targets of diabetes at the intracellular level. This review is dedicated to the analysis of recent data regarding the role of mitochondrial dysfunction in the development of diabetes mellitus. Specific areas of focus include the involvement of mitochondrial calcium transport systems and a pathophysiological phenomenon called the permeability transition pore in the pathogenesis of diabetes mellitus. The important contribution of these systems and their potential relevance as therapeutic targets in the pathology are discussed.
Collapse
Affiliation(s)
- Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Moscow Region, Russia
| | - Natalia V. Belosludtseva
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Moscow Region, Russia
| | - Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Mari El, Russia; (N.V.B.); (M.V.D.)
| |
Collapse
|
18
|
De Marchi U, Fernandez-Martinez S, de la Fuente S, Wiederkehr A, Santo-Domingo J. Mitochondrial ion channels in pancreatic β-cells: Novel pharmacological targets for the treatment of Type 2 diabetes. Br J Pharmacol 2020; 178:2077-2095. [PMID: 32056196 PMCID: PMC8246559 DOI: 10.1111/bph.15018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/18/2022] Open
Abstract
Pancreatic beta‐cells are central regulators of glucose homeostasis. By tightly coupling nutrient sensing and granule exocytosis, beta‐cells adjust the secretion of insulin to the circulating blood glucose levels. Failure of beta‐cells to augment insulin secretion in insulin‐resistant individuals leads progressively to impaired glucose tolerance, Type 2 diabetes, and diabetes‐related diseases. Mitochondria play a crucial role in β‐cells during nutrient stimulation, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion. Mitochondria are double‐membrane organelles containing numerous channels allowing the transport of ions across both membranes. These channels regulate mitochondrial energy production, signalling, and cell death. The mitochondria of β‐cells express ion channels whose physio/pathological role is underappreciated. Here, we describe the mitochondrial ion channels identified in pancreatic β‐cells, we further discuss the possibility of targeting specific β‐cell mitochondrial channels for the treatment of Type 2 diabetes, and we finally highlight the evidence from clinical studies. LINKED ARTICLES This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc
Collapse
Affiliation(s)
| | - Silvia Fernandez-Martinez
- Division of Clinical Pharmacology and Toxicology, Centre de Recherche Clinique, HUG, Genève, Switzerland
| | - Sergio de la Fuente
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
19
|
Taddeo EP, Alsabeeh N, Baghdasarian S, Wikstrom JD, Ritou E, Sereda S, Erion K, Li J, Stiles L, Abdulla M, Swanson Z, Wilhelm JJ, Bellin MD, Kibbey RG, Liesa M, Shirihai OS. Mitochondrial Proton Leak Regulated by Cyclophilin D Elevates Insulin Secretion in Islets at Nonstimulatory Glucose Levels. Diabetes 2020; 69:131-145. [PMID: 31740442 PMCID: PMC6971491 DOI: 10.2337/db19-0379] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Fasting hyperinsulinemia precedes the development of type 2 diabetes. However, it is unclear whether fasting insulin hypersecretion is a primary driver of insulin resistance or a consequence of the progressive increase in fasting glycemia induced by insulin resistance in the prediabetic state. Herein, we have discovered a mechanism that specifically regulates non-glucose-stimulated insulin secretion (NGSIS) in pancreatic islets that is activated by nonesterified free fatty acids, the major fuel used by β-cells during fasting. We show that the mitochondrial permeability transition pore regulator cyclophilin D (CypD) promotes NGSIS, but not glucose-stimulated insulin secretion, by increasing mitochondrial proton leak. Islets from prediabetic obese mice show significantly higher CypD-dependent proton leak and NGSIS compared with lean mice. Proton leak-mediated NGSIS is conserved in human islets and is stimulated by exposure to nonesterified free fatty acids at concentrations observed in obese subjects. Mechanistically, proton leak activates islet NGSIS independently of mitochondrial ATP synthesis but ultimately requires closure of the KATP channel. In summary, we have described a novel nonesterified free fatty acid-stimulated pathway that selectively drives pancreatic islet NGSIS, which may be therapeutically exploited as an alternative way to halt fasting hyperinsulinemia and the progression of type 2 diabetes.
Collapse
Affiliation(s)
- Evan P Taddeo
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Nour Alsabeeh
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Siyouneh Baghdasarian
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jakob D Wikstrom
- Dermatology and Venereology Unit, Department of Medicine, Karolinska Institutet, and Department of Dermato-Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Eleni Ritou
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Samuel Sereda
- Endocrinology, Diabetes, Nutrition and Weight Management Section, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Karel Erion
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jin Li
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Muhamad Abdulla
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Zachary Swanson
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Joshua J Wilhelm
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Melena D Bellin
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Richard G Kibbey
- Departments of Internal Medicine (Endocrinology) and Cellular & Molecular Physiology, Yale University, New Haven, CT
| | - Marc Liesa
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Orian S Shirihai
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
20
|
Hribal ML, Mancuso E, Arcidiacono GP, Greco A, Musca D, Procopio T, Ruffo M, Sesti G. The Phosphatase PHLPP2 Plays a Key Role in the Regulation of Pancreatic Beta-Cell Survival. Int J Endocrinol 2020; 2020:1027386. [PMID: 32411219 PMCID: PMC7199632 DOI: 10.1155/2020/1027386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/03/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
Currently available antidiabetic treatments fail to halt, and may even exacerbate, pancreatic β-cell exhaustion, a key feature of type 2 diabetes pathogenesis; thus, strategies to prevent, or reverse, β-cell failure should be actively sought. The serine threonine kinase Akt has a key role in the regulation of β-cell homeostasis; among Akt modulators, a central role is played by pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) family. Here, taking advantage of an in vitro model of chronic exposure to high glucose, we demonstrated that PHLPPs, particularly the second family member called PHLPP2, are implicated in the ability of pancreatic β cells to deal with glucose toxicity. We observed that INS-1 rat pancreatic β cell line maintained for 12-15 passages at high (30 mM) glucose concentrations (INS-1 HG) showed increased expression of PHLPP2 and PHLPP1 both at mRNA and protein level as compared to INS-1 maintained for the same number of passages in the presence of normal glucose levels (INS-1 NG). These changes were paralleled by decreased phosphorylation of Akt and by increased expression of apoptotic and autophagic markers. To investigate if PHLPPs had a casual role in the alteration of INS-1 homeostasis observed upon chronic exposure to high glucose concentrations, we took advantage of shRNA technology to specifically knock-down PHLPPs. We obtained proof-of-concept evidence that modulating PHLPPs expression may help to restore a healthy β cell mass, as the reduced expression of PHLPP2/1 was accompanied by a recovered balance between pro- and antiapoptotic factor levels. In conclusion, our data provide initial support for future studies aimed to identify pharmacological PHLPPs modulator to treat beta-cell survival impairment. They also contribute to shed some light on β-cell dysfunction, a complex and unsatisfactorily characterized phenomenon that has a central causative role in the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Marta Letizia Hribal
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Elettra Mancuso
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Gaetano Paride Arcidiacono
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
- Department of Medicine, University of Padua, Padua, Italy
| | - Annalisa Greco
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Donatella Musca
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Teresa Procopio
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Mariafrancesca Ruffo
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
- Department of Medicine, Ausl of Bologna, Bologna, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
- Department of Clinical and Molecular Medicine, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
21
|
Ure DR, Trepanier DJ, Mayo PR, Foster RT. Cyclophilin inhibition as a potential treatment for nonalcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2019; 29:163-178. [PMID: 31868526 DOI: 10.1080/13543784.2020.1703948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Daren R. Ure
- Hepion Pharmaceuticals Inc, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
22
|
Li TS, Chen L, Wang SC, Yang YZ, Xu HJ, Gu HM, Zhao XJ, Dong P, Pan Y, Shang ZQ, Zhang XQ, Kong LD. Magnesium isoglycyrrhizinate ameliorates fructose-induced podocyte apoptosis through downregulation of miR-193a to increase WT1. Biochem Pharmacol 2019; 166:139-152. [DOI: 10.1016/j.bcp.2019.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
|
23
|
Ali ES, Petrovsky N. Calcium Signaling As a Therapeutic Target for Liver Steatosis. Trends Endocrinol Metab 2019; 30:270-281. [PMID: 30850262 DOI: 10.1016/j.tem.2019.02.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Hepatic steatosis, the first step in nonalcoholic fatty liver disease (NAFLD), can arise from various pathophysiological conditions. While lipid metabolism in the liver is normally balanced such that there is no excessive lipid accumulation, when this homeostasis is disrupted lipid droplets (LDs) accumulate in hepatocytes resulting in cellular toxicity. The mechanisms underlying this accumulation and the subsequent hepatocellular damage are multifactorial and poorly understood, with the result that there are no currently approved treatments for NAFLD. Impaired calcium signaling has recently been identified as a cause of increased endoplasmic reticulum (ER) stress contributing to hepatic lipid accumulation. This review highlights new findings on the role of impaired Ca2+ signaling in the development of steatosis and discusses potential new approaches to NAFLD treatment based on these new insights.
Collapse
Affiliation(s)
- Eunüs S Ali
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Adelaide, SA, Australia.
| |
Collapse
|
24
|
Faure M, Bertoldo MJ, Khoueiry R, Bongrani A, Brion F, Giulivi C, Dupont J, Froment P. Metformin in Reproductive Biology. Front Endocrinol (Lausanne) 2018; 9:675. [PMID: 30524372 PMCID: PMC6262031 DOI: 10.3389/fendo.2018.00675] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Initially produced in Europe in 1958, metformin is still one of the most widely prescribed drugs to treat type II diabetes and other comorbidities associated with insulin resistance. Metformin has been shown to improve fertility outcomes in females with insulin resistance associated with polycystic ovary syndrome (PCOS) and in obese males with reduced fertility. Metformin treatment reinstates menstrual cyclicity, decreases the incidence of cesareans, and limits the number of premature births. Notably, metformin reduces steroid levels in conditions associated with hyperandrogenism (e.g., PCOS and precocious puberty) in females and improves fertility of adult men with metabolic syndrome through increased testosterone production. While the therapeutical use of metformin is considered to be safe, in the last 10 years some epidemiological studies have described phenotypic differences after prenatal exposure to metformin. The goals of this review are to briefly summarize the current knowledge on metformin focusing on its effects on the female and male reproductive organs, safety concerns, including the potential for modulating fetal imprinting via epigenetics.
Collapse
Affiliation(s)
- Melanie Faure
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - Michael J Bertoldo
- Discipline of Obstetrics and Gynaecology, School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia
| | - Rita Khoueiry
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Alice Bongrani
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - François Brion
- INERIS, Direction des Risques Chroniques, Pole VIVA, Unite d'ecotoxicologie in vitro et in vivo, BP2, Verneuil-en-Halatte, France
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Medical Investigations of Neurodevelopmental Disorders Institute, University of California, Davis, Davis, CA, United States
| | - Joelle Dupont
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| | - Pascal Froment
- Unité de Physiologie de la Reproduction et des Comportements, Centre Val de Loire, Institut National de la Recherche Agronomique, UMR85, Nouzilly, France
| |
Collapse
|
25
|
The protective effect of metformin against the noise-induced hearing loss. Eur Arch Otorhinolaryngol 2018; 275:2957-2966. [DOI: 10.1007/s00405-018-5161-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
|
26
|
Jeon JY, Lee SJ, Lee S, Kim SJ, Han SJ, Kim HJ, Kim DJ, Kim YS, Woo JT, Ahn KJ, Nam M, Baik SH, Park Y, Lee K. Failure of monotherapy in clinical practice in patients with type 2 diabetes: The Korean National Diabetes Program. J Diabetes Investig 2018; 9:1144-1152. [PMID: 29328551 PMCID: PMC6123024 DOI: 10.1111/jdi.12801] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/29/2017] [Accepted: 01/07/2018] [Indexed: 01/20/2023] Open
Abstract
AIMS/INTRODUCTION We investigated the failure of monotherapy in patients with type 2 diabetes mellitus in real practice settings. MATERIALS AND METHODS The Korean National Diabetes Program was a prospective, multicenter observational cohort study of type 2 diabetes mellitus patients in Korea. Of the 3,950 patients enrolled in the study, we studied 998 who were continuously maintained on monotherapy for at least 90 days at six participating centers. To balance the baseline characteristics of patients in each group, we used propensity matching at a 1:1 ratio (metformin vs sulfonylureas) and 4:1 ratio (metformin vs meglitinides and metformin vs alpha-glucosidase inhibitors [aGIs]). The hazard ratios (HRs) of treatments (compared with metformin) were determined by Cox's proportional hazards regression modeling. RESULTS The median follow-up time was 56 months, and monotherapy failed in 45% of all patients. The annual incidences of failure were 15.6%, 21.3%, 27% and 9.6% in the metformin, sulfonylurea, meglitinide and aGI groups. Compared with metformin, sulfonylureas and meglitinides were associated with higher risks of monotherapy failure (HR 1.39, 95% confidence interval [CI] 1.08-1.80; HR 1.92, 95% CI 1.13-3.27), and aGIs with risks similar to that of metformin (HR 0.80, 95% CI 0.44-1.45). When analyzed by failure type, sulfonylureas, meglitinides and aGIs were associated with a higher risk of a switch to other agents (HR 4.43, 95% CI 2.14-9.17; HR 18.80, 95% CI 6.21-56.93; HR 4.25, 95% CI 1.49-12.13), and aGIs with a lower risk of prescription of add-on second agents (HR 0.16, 95% CI 0.04-0.64). CONCLUSIONS Metformin was associated with a lower failure risk than were sulfonylureas and meglitinides, but a comparable aGI failure rate.
Collapse
Affiliation(s)
- Ja Young Jeon
- Department of Endocrinology and MetabolismAjou University School of MedicineSuwonKorea
| | - Soo Jin Lee
- Department of Medicare AdministrationBackseok Arts UniversitySeoulKorea
| | - Sieun Lee
- Office of BiostatisticsAjou University School of MedicineSuwonKorea
| | - Soo Jin Kim
- Office of BiostatisticsAjou University School of MedicineSuwonKorea
| | - Seung Jin Han
- Department of Endocrinology and MetabolismAjou University School of MedicineSuwonKorea
| | - Hae Jin Kim
- Department of Endocrinology and MetabolismAjou University School of MedicineSuwonKorea
| | - Dae Jung Kim
- Department of Endocrinology and MetabolismAjou University School of MedicineSuwonKorea
| | - Young Seol Kim
- Department of Internal MedicineKyung Hee University College of MedicineSeoulKorea
| | - Jeong Taek Woo
- Department of Internal MedicineKyung Hee University College of MedicineSeoulKorea
| | - Kyu Jeung Ahn
- Department of Internal MedicineKyung Hee University College of MedicineSeoulKorea
| | - Moonsuk Nam
- Department of Internal MedicineInha University College of MedicineIncheonKorea
| | - Sei Hyun Baik
- Department of Internal MedicineKorea University College of MedicineSeoulKorea
| | - Yongsoo Park
- Department of Molecular and Integrative PhysiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Kwan‐Woo Lee
- Department of Endocrinology and MetabolismAjou University School of MedicineSuwonKorea
| |
Collapse
|
27
|
Zhou T, Xu X, Du M, Zhao T, Wang J. A preclinical overview of metformin for the treatment of type 2 diabetes. Biomed Pharmacother 2018; 106:1227-1235. [PMID: 30119191 DOI: 10.1016/j.biopha.2018.07.085] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/14/2018] [Accepted: 07/15/2018] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes (T2D) is the most common type of diabetes mellitus and is mainly characterized by insulin resistance, β-cell dysfunction, and elevated hepatic glucose output. Metformin is a first-line antihyperglycemic agent that works mainly by regulating hepatic glucose production and peripheral insulin sensitivity. Metformin has been clinically applied for more than half a century, although the underlying pharmacological mechanisms remain elusive. This current review mainly focused on the development history of metformin and related preclinical studies on structural modification, pharmacological mechanisms for treatment of T2D, toxicology, pharmacokinetics, and pharmaceutics. The pharmacological function of metformin in lowering hyperglycemia suggests that multi-targeting could be an effective strategy for the discovery of new anti-diabetic drugs. A number of discoveries have revealed the pharmacologic mechanisms of metformin; however, precise mechanisms remain unclear. Deeper investigations on the biological features of metformin are expected to provide more rational applications and indications of this evergreen anti-T2D agent, which will in turn help to better understand the complicated pathogenesis of T2D.
Collapse
Affiliation(s)
- Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; Shanghai Institute of Material Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Xin Xu
- Shanghai Institute of Material Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Mengfan Du
- Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Tong Zhao
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Jiaying Wang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China.
| |
Collapse
|
28
|
Fontaine E. Metformin-Induced Mitochondrial Complex I Inhibition: Facts, Uncertainties, and Consequences. Front Endocrinol (Lausanne) 2018; 9:753. [PMID: 30619086 PMCID: PMC6304344 DOI: 10.3389/fendo.2018.00753] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/27/2018] [Indexed: 12/24/2022] Open
Abstract
Metformin is the most widely prescribed drug to treat patients with type II diabetes, for whom retrospective studies suggest that metformin may have anticancer properties. However, in experiments performed with isolated cells, authors have reported both pro- and anti-apoptotic effects of metformin. The exact molecular mechanism of action of metformin remains partly unknown despite its use for over 60 years and more than 17,000 articles in PubMed. Among the various widely recognized or recently proposed targets, it has been reported consistently that metformin is capable of inhibiting mitochondrial respiratory chain Complex I. Since most of the effects of metformin have been replicated by other inhibitors of Complex I, it has been suggested that the mechanism of action of metformin involved the inhibition of Complex I. However, compared to conventional Complex I inhibitors, the metformin-induced inhibition of Complex I has unique characteristics. Among these, the most original one is that the concentrations of metformin required to inhibit Complex I are lower in intact cells than in isolated mitochondria. Experiments with isolated mitochondria or Complex I were generally performed using millimolar concentrations of metformin, while plasma levels remain in the micromolar range in both human and animal studies, highlighting that metformin concentration is an important issue. In order to explain the effects in animals based on observations in cells and mitochondria, some authors proposed a direct effect of the drug on Complex I involving an accumulation of metformin inside the mitochondria while others proposed an indirect effect (the drug no longer having to diffuse into the mitochondria). This brief review attempts to: gather arguments for and against each hypothesis concerning the mechanism by which metformin inhibits Complex I and to highlight remaining questions about the toxicity mechanism of metformin for certain cancer cells.
Collapse
|
29
|
Lamarche F, Cottet-Rousselle C, Barret L, Fontaine E. Protection of PC12 cells from cocaine-induced cell death by inhibiting mitochondrial permeability transition. Neurochem Int 2017; 109:34-40. [DOI: 10.1016/j.neuint.2017.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/21/2022]
|
30
|
Abstract
The pancreatic β-cell secretes insulin in response to elevated plasma glucose. This review applies an external bioenergetic critique to the central processes of glucose-stimulated insulin secretion, including glycolytic and mitochondrial metabolism, the cytosolic adenine nucleotide pool, and its interaction with plasma membrane ion channels. The control mechanisms responsible for the unique responsiveness of the cell to glucose availability are discussed from bioenergetic and metabolic control standpoints. The concept of coupling factor facilitation of secretion is critiqued, and an attempt is made to unravel the bioenergetic basis of the oscillatory mechanisms controlling secretion. The need to consider the physiological constraints operating in the intact cell is emphasized throughout. The aim is to provide a coherent pathway through an extensive, complex, and sometimes bewildering literature, particularly for those unfamiliar with the field.
Collapse
Affiliation(s)
- David G Nicholls
- Buck Institute for Research on Aging, Novato, California; and Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmo, Sweden
| |
Collapse
|
31
|
Safi SZ, Qvist R, Ong G, Karimian H, Imran M, Shah I. Stimulation of β-adrenergic receptors plays a protective role via increased expression of RAF-1 and PDX-1 in hyperglycemic rat pancreatic islet (RIN-m5F) cells. Arch Med Sci 2017; 13:470-480. [PMID: 28261303 PMCID: PMC5332455 DOI: 10.5114/aoms.2016.64131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 07/14/2015] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION It is a widely held view that a progressive reduction of beta-cell mass occurs in the progression of diabetes. RAF-1 kinase and pancreas duodenal homeobox 1 (PDX-1) are major factors that promote survival of cells and maintain normal insulin functions. In this study we investigated the effect of a β-adrenergic receptor agonist and antagonist on RAF-1 and PDX-1, and their respective effects on apoptosis and insulin release in RIN-m5F cells. MATERIAL AND METHODS RIN-m5F cells were cultured in normal (5 mM) and high (25 mM) glucose to mimic diabetic conditions, followed by treatment with 5 µM, 10 µM and 20 µM of isoproterenol and isoproterenol + propranolol for 6, 12 and 24 h. Western blotting and reverse transcription analysis were performed to examine the expression of RAF-1 and PDX-1. Annexin-V-FITC and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assays were used to investigate apoptosis. ELISA was used to measure insulin levels. Reverse transcription polymerase chain reaction was conducted to investigate the expression of genes. RESULTS Stimulation of β-adrenergic receptors with isoproterenol significantly induced RAF-1 and PDX-1 genes in a concentration-dependent and time-independent manner. Changes were significant both at protein and mRNA levels. Up-regulation of RAF-1 and PDX-1 was accompanied by improved insulin levels and reduced apoptosis. Concentrations of 10 µM and 20 µM for 12 and 24 h were more effective in achieving significant differences in the experimental and control groups. Propranolol reversed the effect of isoproterenol mostly at maximum concentrations and time periods. CONCLUSIONS A positive effect of a β-adrenergic agonist on RAF-1 and PDX-1, reduction in β-cell apoptosis and improved insulin contents can help to understand the pathogenesis of diabetes and to develop novel approaches for the β-cell dysfunction in diabetes.
Collapse
Affiliation(s)
- Sher Zaman Safi
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology, Lahore, Pakistan
| | - Rajes Qvist
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Gracie Ong
- Department of Anesthesiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Muhammad Imran
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Ikram Shah
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Yang X, Xu Z, Zhang C, Cai Z, Zhang J. Metformin, beyond an insulin sensitizer, targeting heart and pancreatic β cells. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1984-1990. [PMID: 27702625 DOI: 10.1016/j.bbadis.2016.09.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/17/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022]
Abstract
Metformin, a biguanide derivate, is known as the first-line antidiabetic agent for type 2 diabetes mellitus (T2DM) treatment. It reduces insulin resistance and decreases blood glucose concentration by inhibiting gluconeogenesis and suppressing hepatic glucose production with improved peripheral tissue insulin sensitivity. As an insulin sensitizer, metformin takes pleiotropic actions and exerts protective effects on multiple organs mainly in insulin-targeted tissues such as liver, muscle, and adipose tissues. Recent studies discover that metformin also plays essential roles in heart and pancreatic β cells - two important organs in metabolic regulation. Metformin not only protects T2DM patients from cardiovascular diseases and heart failure, but also restores insulin secretion activities and protects pancreatic β cells from lipotoxicity or glucotoxicity. Although accumulated evidence shed light on the metformin action, the precise mechanism of metformin is still under investigation. Further laboratory investigations and clinical trials are needed to pinpoint a map of metformin action. Based on recent findings, this review characterizes the beneficial role of metformin in cardiovascular diseases and pancreatic β cells.
Collapse
Affiliation(s)
- Xin Yang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| | - Zhipeng Xu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| | - Chunlan Zhang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| | - Zixin Cai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China
| | - Jingjing Zhang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
33
|
TRPV6 modulates proliferation of human pancreatic neuroendocrine BON-1 tumour cells. Biosci Rep 2016; 36:BSR20160106. [PMID: 27450545 PMCID: PMC4995500 DOI: 10.1042/bsr20160106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/22/2016] [Indexed: 11/27/2022] Open
Abstract
Highly Ca2+ permeable receptor potential channel vanilloid type 6 (TRPV6) modulates a variety of biological functions including calcium-dependent cell growth and apoptosis. So far, the role of TRPV6 in controlling growth of pancreatic neuroendocrine tumour (NET) cells is unknown. In the present study, we characterize the expression of TRPV6 in pancreatic BON-1 and QGP-1 NET cells. Furthermore, we evaluate the impact of TRPV6 on intracellular calcium, the activity of nuclear factor of activated T-cells (NFAT) and proliferation of BON-1 cells. TRPV6 expression was assessed by real-time PCR and Western blot. TRPV6 mRNA expression and protein production were down-regulated by siRNA. Changes in intracellular calcium levels were detected by fluorescence calcium imaging (fura-2/AM). NFAT activity was studied by NFAT reporter assay; cell proliferation by bromodeoxyuridine (BrdU), MTT and propidium iodine staining. TRPV6 mRNA and protein are present in BON-1 and QGP-1 NET-cells. Down-regulation of TRPV6 attenuates BON-1 cell proliferation. TRPV6 down-regulation is associated with decreased Ca2+ response pattern and reduced NFAT activity. In conclusion, TRPV6 is expressed in pancreatic NETs and modulates cell proliferation via Ca2+-dependent mechanism, which is accompanied by NFAT activation.
Collapse
|
34
|
Yang Y, Chan L. Monogenic Diabetes: What It Teaches Us on the Common Forms of Type 1 and Type 2 Diabetes. Endocr Rev 2016; 37:190-222. [PMID: 27035557 PMCID: PMC4890265 DOI: 10.1210/er.2015-1116] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To date, more than 30 genes have been linked to monogenic diabetes. Candidate gene and genome-wide association studies have identified > 50 susceptibility loci for common type 1 diabetes (T1D) and approximately 100 susceptibility loci for type 2 diabetes (T2D). About 1-5% of all cases of diabetes result from single-gene mutations and are called monogenic diabetes. Here, we review the pathophysiological basis of the role of monogenic diabetes genes that have also been found to be associated with common T1D and/or T2D. Variants of approximately one-third of monogenic diabetes genes are associated with T2D, but not T1D. Two of the T2D-associated monogenic diabetes genes-potassium inward-rectifying channel, subfamily J, member 11 (KCNJ11), which controls glucose-stimulated insulin secretion in the β-cell; and peroxisome proliferator-activated receptor γ (PPARG), which impacts multiple tissue targets in relation to inflammation and insulin sensitivity-have been developed as major antidiabetic drug targets. Another monogenic diabetes gene, the preproinsulin gene (INS), is unique in that INS mutations can cause hyperinsulinemia, hyperproinsulinemia, neonatal diabetes mellitus, one type of maturity-onset diabetes of the young (MODY10), and autoantibody-negative T1D. Dominant heterozygous INS mutations are the second most common cause of permanent neonatal diabetes. Moreover, INS gene variants are strongly associated with common T1D (type 1a), but inconsistently with T2D. Variants of the monogenic diabetes gene Gli-similar 3 (GLIS3) are associated with both T1D and T2D. GLIS3 is a key transcription factor in insulin production and β-cell differentiation during embryonic development, which perturbation forms the basis of monogenic diabetes as well as its association with T1D. GLIS3 is also required for compensatory β-cell proliferation in adults; impairment of this function predisposes to T2D. Thus, monogenic forms of diabetes are invaluable "human models" that have contributed to our understanding of the pathophysiological basis of common T1D and T2D.
Collapse
Affiliation(s)
- Yisheng Yang
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Lawrence Chan
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
35
|
Millet A, Bouzat P, Trouve-Buisson T, Batandier C, Pernet-Gallay K, Gaide-Chevronnay L, Barbier EL, Debillon T, Fontaine E, Payen JF. Erythropoietin and Its Derivates Modulate Mitochondrial Dysfunction after Diffuse Traumatic Brain Injury. J Neurotrauma 2016; 33:1625-33. [PMID: 26530102 DOI: 10.1089/neu.2015.4160] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Inhibiting the opening of mitochondrial permeability transition pore (mPTP), thereby maintaining the mitochondrial membrane potential and calcium homeostasis, could reduce the induction of cell death. Although recombinant human erythropoietin (rhEpo) and carbamylated erythropoietin (Cepo) were shown to prevent apoptosis after traumatic brain injury (TBI), their impact on mPTP is yet unknown. Thirty minutes after diffuse TBI (impact-acceleration model), rats were intravenously administered a saline solution (TBI-saline), 5000 UI/kg rhEpo (TBI-rhEpo) or 50 μg/kg Cepo (TBI-Cepo). A fourth group received no TBI insult (sham-operated) (n = 11 rats per group). Post-traumatic brain edema was measured using magnetic resonance imaging. A first series of experiments was conducted 2 h after TBI (or equivalent) to investigate the mitochondrial function with the determination of thresholds for mPTP opening and ultrastructural mitochondrial changes. In addition, the intramitochondrial calcium content [Caim] was measured. In a second series of experiments, brain cell apoptosis was assessed at 24 h post-injury. TBI-rhEpo and TBI-Cepo groups had a reduced brain edema compared with TBI-saline. They had higher threshold for mPTP opening with succinate as substrate: 120 (120-150) (median, interquartiles) and 100 (100-120) versus 80 (60-90) nmol calcium/mg protein in TBI-saline, respectively (p < 0.05). Similar findings were shown with glutamate-malate as substrate. TBI-rhEpo and Cepo groups had less morphological mitochondrial disruption in astrocytes. The elevation in [Caim] after TBI was not changed by rhEpo and Cepo treatment. Finally, rhEpo and Cepo reduced caspase-3 expression at 24 h post-injury. These results indicate that rhEpo and Cepo could modulate mitochondrial dysfunction after TBI. The mechanisms involved are discussed.
Collapse
Affiliation(s)
- Anne Millet
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France .,3 Département de Réanimation Pédiatrique et Néonatale, Hôpital Couple Enfant , Grenoble, France
| | - Pierre Bouzat
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France .,4 Pôle d'Anesthésie Réanimation, CHU Grenoble Alpes , Grenoble, France
| | - Thibaut Trouve-Buisson
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France .,4 Pôle d'Anesthésie Réanimation, CHU Grenoble Alpes , Grenoble, France
| | - Cécile Batandier
- 5 INSERM, U1055, Laboratoire de Biologie Fondamentale et Appliquée, Université Joseph Fourier , Grenoble, France
| | - Karin Pernet-Gallay
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France
| | - Lucie Gaide-Chevronnay
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France .,4 Pôle d'Anesthésie Réanimation, CHU Grenoble Alpes , Grenoble, France
| | | | - Thierry Debillon
- 3 Département de Réanimation Pédiatrique et Néonatale, Hôpital Couple Enfant , Grenoble, France
| | - Eric Fontaine
- 5 INSERM, U1055, Laboratoire de Biologie Fondamentale et Appliquée, Université Joseph Fourier , Grenoble, France .,6 Unité de Nutrition Parentérale, Pôle de médecin Aigue Spécialisée, CHU Grenoble Alpes , Grenoble, France
| | - Jean-François Payen
- 1 INSERM , U1216, Grenoble, France .,2 Grenoble Institut des Neurosciences, Université Grenoble Alpes , Grenoble, France .,4 Pôle d'Anesthésie Réanimation, CHU Grenoble Alpes , Grenoble, France
| |
Collapse
|
36
|
Imeglimin prevents human endothelial cell death by inhibiting mitochondrial permeability transition without inhibiting mitochondrial respiration. Cell Death Discov 2016; 2:15072. [PMID: 27551496 PMCID: PMC4979505 DOI: 10.1038/cddiscovery.2015.72] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 11/22/2015] [Indexed: 02/07/2023] Open
Abstract
Imeglimin is the first in a new class of oral glucose-lowering agents, having recently completed its phase 2b trial. As Imeglimin did show a full prevention of β-cell apoptosis, and since angiopathy represents a major complication of diabetes, we studied Imeglimin protective effects on hyperglycemia-induced death of human endothelial cells (HMEC-1). These cells were incubated in several oxidative stress environments (exposure to high glucose and oxidizing agent tert-butylhydroperoxide) which led to mitochondrial permeability transition pore (PTP) opening, cytochrome c release and cell death. These events were fully prevented by Imeglimin treatment. This protective effect on cell death occurred without any effect on oxygen consumption rate, on lactate production and on cytosolic redox or phosphate potentials. Imeglimin also dramatically decreased reactive oxygen species production, inhibiting specifically reverse electron transfer through complex I. We conclude that Imeglimin prevents hyperglycemia-induced cell death in HMEC-1 through inhibition of PTP opening without inhibiting mitochondrial respiration nor affecting cellular energy status. Considering the high prevalence of macrovascular and microvascular complications in type 2 diabetic subjects, these results together suggest a potential benefit of Imeglimin in diabetic angiopathy.
Collapse
|
37
|
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
|
38
|
Glutz A, Leitmeyer K, Setz C, Brand Y, Bodmer D. Metformin Protects Auditory Hair Cells from Gentamicin-Induced Toxicity in vitro. Audiol Neurootol 2015; 20:360-9. [PMID: 26372952 DOI: 10.1159/000438918] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 07/23/2015] [Indexed: 11/19/2022] Open
Abstract
Metformin is a commonly used antidiabetic drug. It has been shown that this drug activates the AMP-activated protein kinase, which inhibits downstream the mammalian target of rapamycin. In addition, several studies indicate that metformin reduces intracellular reactive oxygen species. Our data, using an in vitro rat model, indicate that metformin is able to protect auditory hair cells (HCs) from gentamicin-induced apoptotic cell death. Moreover, metformin has no toxic effect on spiral ganglion neuronal survival or outgrowth in vitro. These results suggest a protective effect of metformin on auditory HC survival in gentamicin-induced HC loss in vitro.
Collapse
Affiliation(s)
- Andrea Glutz
- Department of Biomedicine, Head and Neck Surgery, University Hospital Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
39
|
Raúl BG, Antonio FLL, Arturo BGL, Miguel C, Rebeca GM, Alejandro ÁR, Alejandra CR, Margarita DF, Clara OC. Hyperglycemia promotes p53-Mdm2 interaction but reduces p53 ubiquitination in RINm5F cells. Mol Cell Biochem 2015; 405:257-64. [DOI: 10.1007/s11010-015-2416-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/18/2015] [Indexed: 12/31/2022]
|
40
|
Lablanche S, Cottet-Rousselle C, Argaud L, Laporte C, Lamarche F, Richard MJ, Berney T, Benhamou PY, Fontaine E. Respective effects of oxygen and energy substrate deprivation on beta cell viability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:629-39. [PMID: 25868875 DOI: 10.1016/j.bbabio.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/30/2015] [Accepted: 04/05/2015] [Indexed: 12/25/2022]
Abstract
Deficit in oxygen and energetic substrates delivery is a key factor in islet loss during islet transplantation. Permeability transition pore (PTP) is a mitochondrial channel involved in cell death. We have studied the respective effects of oxygen and energy substrate deprivation on beta cell viability as well as the involvement of oxidative stress and PTP opening. Energy substrate deprivation for 1h followed by incubation in normal conditions led to a cyclosporin A (CsA)-sensitive-PTP-opening in INS-1 cells and human islets. Such a procedure dramatically decreased INS-1 cells viability except when transient removal of energy substrates was performed in anoxia, in the presence of antioxidant N-acetylcysteine (NAC) or when CsA or metformin inhibited PTP opening. Superoxide production increased during removal of energy substrates and increased again when normal energy substrates were restored. NAC, anoxia or metformin prevented the two phases of oxidative stress while CsA prevented the second one only. Hypoxia or anoxia alone did not induce oxidative stress, PTP opening or cell death. In conclusion, energy substrate deprivation leads to an oxidative stress followed by PTP opening, triggering beta cell death. Pharmacological prevention of PTP opening during islet transplantation may be a suitable option to improve islet survival and graft success.
Collapse
Affiliation(s)
- Sandrine Lablanche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France.
| | - Cécile Cottet-Rousselle
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | | | - Camille Laporte
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Frédéric Lamarche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Marie-Jeanne Richard
- Cellular Therapy Unit, EFS Rhône-Alpes, Grenoble University Hospital, Grenoble, France
| | - Thierry Berney
- Cell Isolation and Transplant Center, University of Geneva, Level R, 1 rue Michel Servet, Geneva 4, CH-1211, Switzerland
| | - Pierre-Yves Benhamou
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| | - Eric Fontaine
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| |
Collapse
|
41
|
Chandravanshi B, Dhanushkodi A, Bhonde R. High Recovery of Functional Islets Stored at Low and Ultralow Temperatures. Rev Diabet Stud 2015; 11:267-78. [PMID: 26177487 DOI: 10.1900/rds.2014.11.267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Poor recovery of islets upon cryopreservation is the main hurdle in islet banking. Pancreatic islets have a poor antioxidative defense mechanism, and exposure of islets to low temperature leads to oxidative stress. AIM We aimed to investigate whether known compounds such as metformin, γ aminobutyric acid (GABA), docosahexanoic acid (DHA), or eicosapentaenoic acid (EPA) alone or in combination are capable of reducing oxidative stress for better islet recovery upon storage at suboptimal temperatures. METHODS Islets isolated from mouse pancreas were stored at low temperature (4°C) for 15 days and at ultralow temperature (-196°C) for 30 days with or without additives. After revival from cold storage, islets were assessed by using three methods: (1) specificity by dithizone (DTZ), (2) viability by fluorescein diacetate/propidium iodide (FDA/PI) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-zolium bromide (MTT) assay, and (3) functionality by glucose-stimulated insulin secretion (GSIS). The oxidative status of the islets stored at suboptimal temperatures was determined by both intracellular free radical release (fluorometric analysis) and lipid peroxidation (enzymatic determination). RESULTS Supplementation with additives led to an improvement in islet survival upon storage at suboptimal temperatures, without depletion of insulin secretory activity, which was comparable to that of controls. The additives acted as cryoprotectants and antioxidants as revealed by high recovery of viable islets and reduction in total reactive oxygen species (ROS) and malonidealdehyde (MDA), respectively. CONCLUSIONS Our results demonstrate for the first time that supplementation with EPA, DHA, and metformin may lead to higher islet recovery from -196°C storage, enabling proper islet banking.
Collapse
Affiliation(s)
- Bhawna Chandravanshi
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| | - Anandh Dhanushkodi
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| | - Ramesh Bhonde
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| |
Collapse
|
42
|
Batandier C, Poulet L, Hininger I, Couturier K, Fontaine E, Roussel AM, Canini F. Acute stress delays brain mitochondrial permeability transition pore opening. J Neurochem 2014; 131:314-22. [PMID: 24989320 DOI: 10.1111/jnc.12811] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/28/2014] [Accepted: 06/28/2014] [Indexed: 12/15/2022]
Abstract
Since emotional stress elicits brain activation, mitochondria should be a key component of stressed brain response. However, few studies have focused on mitochondria functioning in these conditions. In this work, we aimed to determine the effects of an acute restraint stress on rat brain mitochondrial functions, with a focus on permeability transition pore (PTP) functioning. Rats were divided into two groups, submitted or not to an acute 30-min restraint stress (Stress, S-group, vs. Control, C-group). Brain was removed immediately after stress. Mitochondrial respiration and enzymatic activities (complex I, complex II, hexokinase) were measured. Changes in PTP opening were assessed by the Ca(2+) retention capacity. Cell signaling pathways relevant to the coupling between mitochondria and cell function (adenosine monophosphate-activated protein kinase, phosphatidylinositol 3-kinase, glycogen synthase kinase 3 beta, MAPK, and cGMP/NO) were measured. The effect of glucocorticoids was also assessed in vitro. Stress delayed (43%) the opening of PTP and resulted in a mild inhibition of complex I respiratory chain. This inhibition was associated with significant stress-induced changes in adenosine monophosphate-activated protein kinase signaling pathway without changes in brain cGMP level. In contrast, glucocorticoids did not modify PTP opening. These data suggest a rapid adaptive mechanism of brain mitochondria in stressed conditions, with a special focus on PTP regulation. In a rat model of acute restraint stress, we observed substantial changes in brain mitochondria functioning. Stress significantly (i) delays (43%) the opening of permeability transition pore (PTP) by the calcium (Ca(2+) ), its main inductor and (ii) results in an inhibition of complex I in electron transport chain associated with change in AMPK signaling pathway. These data suggest an adaptive mechanism of brain mitochondria in stressed condition, with a special focus on PTP regulation.
Collapse
Affiliation(s)
- Cécile Batandier
- Laboratoire de Bioénergétique Fondamentale et Appliquée, Université Joseph Fourier, Grenoble, France; U1055 - INSERM, Grenoble, France
| | | | | | | | | | | | | |
Collapse
|
43
|
Gutiérrez-Aguilar M, Douglas DL, Gibson AK, Domeier TL, Molkentin JD, Baines CP. Genetic manipulation of the cardiac mitochondrial phosphate carrier does not affect permeability transition. J Mol Cell Cardiol 2014; 72:316-25. [PMID: 24768964 PMCID: PMC4080311 DOI: 10.1016/j.yjmcc.2014.04.008] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 04/10/2014] [Accepted: 04/13/2014] [Indexed: 02/07/2023]
Abstract
The Mitochondrial Permeability Transition (MPT) pore is a voltage-sensitive unselective channel known to instigate necrotic cell death during cardiac disease. Recent models suggest that the isomerase cyclophilin D (CypD) regulates the MPT pore by binding to either the F0F1-ATP synthase lateral stalk or the mitochondrial phosphate carrier (PiC). Here we confirm that CypD, through its N-terminus, can directly bind PiC. We then generated cardiac-specific mouse strains overexpressing or with decreased levels of mitochondrial PiC to assess the functionality of such interaction. While PiC overexpression had no observable pathologic phenotype, PiC knockdown resulted in cardiac hypertrophy along with decreased ATP levels. Mitochondria isolated from the hearts of these mouse lines and their respective non-transgenic controls had no divergent phenotype in terms of oxygen consumption and Ca(2+)-induced MPT, as assessed by swelling and Ca(2+)-retention measurements. These results provide genetic evidence indicating that the mitochondrial PiC is not a critical component of the MPT pore.
Collapse
Affiliation(s)
| | - Diana L Douglas
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Anne K Gibson
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA; Howard Hughes Medical Institute, Cincinnati, OH 45229, USA
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO 65211, USA; Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO 65211, USA.
| |
Collapse
|
44
|
Vogt JA, Wachter U, Wagner K, Calzia E, Gröger M, Weber S, Stahl B, Georgieff M, Asfar P, Fontaine E, Radermacher P, Leverve XM, Wagner F. Effects of glycemic control on glucose utilization and mitochondrial respiration during resuscitated murine septic shock. Intensive Care Med Exp 2014; 2:19. [PMID: 26266919 PMCID: PMC4678133 DOI: 10.1186/2197-425x-2-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/08/2014] [Indexed: 12/13/2022] Open
Abstract
Background This study aims to test the hypothesis whether lowering glycemia improves mitochondrial function and thereby attenuates apoptotic cell death during resuscitated murine septic shock. Methods Immediately and 6 h after cecal ligation and puncture (CLP), mice randomly received either vehicle or the anti-diabetic drug EMD008 (100 μg · g-1). At 15 h post CLP, mice were anesthetized, mechanically ventilated, instrumented and rendered normo- or hyperglycemic (target glycemia 100 ± 20 and 180 ± 50 mg · dL-1, respectively) by infusing stable, non-radioactive isotope-labeled 13C6-glucose. Target hemodynamics was achieved by colloid fluid resuscitation and continuous i.v. noradrenaline, and mechanical ventilation was titrated according to blood gases and pulmonary compliance measurements. Gluconeogenesis and glucose oxidation were derived from blood and expiratory glucose and 13CO2 isotope enrichments, respectively; mathematical modeling allowed analyzing isotope data for glucose uptake as a function of glycemia. Postmortem liver tissue was analyzed for HO-1, AMPK, caspase-3, and Bax (western blotting) expression as well as for mitochondrial respiratory activity (high-resolution respirometry). Results Hyperglycemia lowered mitochondrial respiratory capacity; EMD008 treatment was associated with increased mitochondrial respiration. Hyperglycemia decreased AMPK phosphorylation, and EMD008 attenuated both this effect as well as the expression of activated caspase-3 and Bax. During hyperglycemia EMD008 increased HO-1 expression. During hyperglycemia, maximal mitochondrial oxidative phosphorylation rate was directly related to HO-1 expression, while it was unrelated to AMPK activation. According to the mathematical modeling, EMD008 increased the slope of glucose uptake plotted as a function of glycemia. Conclusions During resuscitated, polymicrobial, murine septic shock, glycemic control either by reducing glucose infusion rates or EMD008 improved glucose uptake and thereby liver tissue mitochondrial respiratory activity. EMD008 effects were more pronounced during hyperglycemia and coincided with attenuated markers of apoptosis. The effects of glucose control were at least in part due to the up-regulation of HO-1 and activation of AMPK. Electronic supplementary material The online version of this article (doi:10.1186/2197-425X-2-19) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Josef A Vogt
- Sektion Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Klinik für Anästhesiologie, Universitätsklinikum, Helmhotzstrasse 8-1, Ulm, 89081, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Masini M, Anello M, Bugliani M, Marselli L, Filipponi F, Boggi U, Purrello F, Occhipinti M, Martino L, Marchetti P, De Tata V. Prevention by metformin of alterations induced by chronic exposure to high glucose in human islet beta cells is associated with preserved ATP/ADP ratio. Diabetes Res Clin Pract 2014; 104:163-70. [PMID: 24462282 DOI: 10.1016/j.diabres.2013.12.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 12/13/2013] [Accepted: 12/21/2013] [Indexed: 11/25/2022]
Abstract
AIM We have explored whether the insulin secretory defects induced by glucotoxicity in human pancreatic islets could be prevented by metformin and investigated some of the possible mechanisms involved. METHODS Human pancreatic islets and INS-1E cells were cultured for 24h with or without high glucose (16.7mM) concentration in the presence or absence of therapeutical concentration of metformin and then glucose-stimulated insulin release, adenine nucleotide levels and mitochondrial complex I and II activities were measured. Islet ultrastructure was analyzed by electron microscopy. RESULTS Compared to control islets, human islets cultured with high glucose showed a reduced glucose-stimulated insulin secretion that was associated with lower ATP levels and a lower ATP/ADP ratio. These functional and biochemical defects were significantly prevented by the presence of metformin in the culture medium, that was also able to significantly inhibit the activity of mitochondrial complex I especially in beta cells exposed to high glucose. Ultrastructural observations showed that mitochondrial volume density was significantly increased in high glucose cultured islets. The critical involvement of mitochondria was further supported by the observation of remarkably swollen organelles with dispersed matrix and fragmented cristae. Metformin was able to efficiently prevent the appearance of all these ultrastructural alterations in human islets exposed to high glucose. CONCLUSIONS Our results show that the functional, biochemical and ultrastructural abnormalities observed in human islet cells exposed to glucotoxic condition can be significantly prevented by metformin, further highlighting a direct beneficial effect of this drug on the insulin secreting human pancreatic beta cells.
Collapse
Affiliation(s)
- M Masini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - M Anello
- Department of Clinical and Molecular Biomedicine, University of Catania, Italy
| | - M Bugliani
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - L Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - F Filipponi
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Italy
| | - U Boggi
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Italy
| | - F Purrello
- Department of Clinical and Molecular Biomedicine, University of Catania, Italy
| | - M Occhipinti
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - L Martino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - P Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - V De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy.
| |
Collapse
|
46
|
Jiang Y, Huang W, Wang J, Xu Z, He J, Lin X, Zhou Z, Zhang J. Metformin plays a dual role in MIN6 pancreatic β cell function through AMPK-dependent autophagy. Int J Biol Sci 2014; 10:268-77. [PMID: 24644425 PMCID: PMC3957082 DOI: 10.7150/ijbs.7929] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/07/2014] [Indexed: 12/20/2022] Open
Abstract
Metformin improves insulin sensitivity in insulin sensitive tissues such as liver, muscle and fat. However, the functional roles and the underlying mechanism of metformin action in pancreatic β cells remain elusive. Here we show that, under normal growth condition, metformin suppresses MIN6 β cell proliferation and promotes apoptosis via an AMPK-dependent and autophagy-mediated mechanism. On the other hand, metformin protects MIN6 cells against palmitic acid (PA)-induced apoptosis. Our findings indicate that metformin plays a dual role in β cell survival and overdose of this anti-diabetic drug itself may lead to potential β cell toxicity.
Collapse
Affiliation(s)
- Yingling Jiang
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011 ; 2. Endocrinology Department, Zhuzhou Central Hospital, Zhuzhou, Hunan, China 412007
| | - Wei Huang
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Jing Wang
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Zhipeng Xu
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Jieyu He
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Xiaohong Lin
- 2. Endocrinology Department, Zhuzhou Central Hospital, Zhuzhou, Hunan, China 412007
| | - Zhiguang Zhou
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| | - Jingjing Zhang
- 1. Metabolic Syndrome Research Center and Diabetes Center, Institute of Aging and Geriatric Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China 410011
| |
Collapse
|
47
|
Vetere A, Choudhary A, Burns SM, Wagner BK. Targeting the pancreatic β-cell to treat diabetes. Nat Rev Drug Discov 2014; 13:278-89. [PMID: 24525781 DOI: 10.1038/nrd4231] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes is a leading cause of morbidity and mortality worldwide, and predicted to affect over 500 million people by 2030. However, this growing burden of disease has not been met with a comparable expansion in therapeutic options. The appreciation of the pancreatic β-cell as a central player in the pathogenesis of both type 1 and type 2 diabetes has renewed focus on ways to improve glucose homeostasis by preserving, expanding and improving the function of this key cell type. Here, we provide an overview of the latest developments in this field, with an emphasis on the most promising strategies identified to date for treating diabetes by targeting the β-cell.
Collapse
Affiliation(s)
- Amedeo Vetere
- Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Amit Choudhary
- 1] Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA. [2] Society of Fellows, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Sean M Burns
- Medical & Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Bridget K Wagner
- Chemical Biology Program, Center for the Science of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
48
|
Ramachandran S, Venugopal A, Kutty VR, A V, G D, Chitrasree V, Mullassari A, Pratapchandran NS, Santosh KR, Pillai MR, Kartha CC. Plasma level of cyclophilin A is increased in patients with type 2 diabetes mellitus and suggests presence of vascular disease. Cardiovasc Diabetol 2014; 13:38. [PMID: 24502618 PMCID: PMC3922405 DOI: 10.1186/1475-2840-13-38] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/05/2014] [Indexed: 11/30/2022] Open
Abstract
Aims/hypothesis Cyclophilin A, an immunophilin is secreted from human monocytes activated by high glucose. Given its role as an inflammatory mediator of vascular tissue damage associated with inflammation and oxidative stress, we examined plasma levels of cyclophilin A in normal healthy volunteers and patients with type 2 diabetes (DM), with or without coronary artery disease (CAD). Methods Study subjects comprised of 212 patients with DM and CAD,101 patients with diabetes, 122 patients with CAD and 121 normal healthy volunteers. Diabetes was assessed by HbA1c levels while coronary artery disease was established by a positive treadmill test and/or coronary angiography. Plasma cyclophilin A was measured using a cyclophilin A ELISA Kit. Relationship of plasma cyclophilin A levels with blood markers of type 2 diabetes, blood lipid levels and medication for diabetes and coronary artery disease were also explored. Results Plasma Cyclophilin levels were higher in diabetes patients with or without CAD compared to normal subjects (P < 0.001). Age, fasting blood sugar levels and HbA1C levels were positively associated with increased plasma cyclophilin. Patients using metformin had reduced levels of plasma cyclophilin (p < 0.001).Serum levels of total cholesterol, LDL cholesterol and triglycerides had no significant association with plasma cyclophilin levels. In patients with increased serum CRP levels, plasma cyclophilin A was also elevated (p = 0.016). Prevalence odds for DM, DM + CAD and CAD are higher in those with high cyclophilin values, compared to those with lower values, after adjusting for age and sex, indicating strong association of high cyclophilin values with diabetes and vascular disease. Conclusions/interpretations Our study demonstrates that patients with type 2 diabetes have higher circulating levels of cyclophilin A than the normal population. Plasma cyclophilin levels were increased in patients with diabetes and coronary artery disease suggesting a role of this protein in accelerating vascular disease in type 2 diabetes. Considering the evidence that Cyclophilin A is an inflammatory mediator in atherogenesis, the mechanistic role of cyclophilin A in diabetic vascular disease progression deserves detailed investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - C C Kartha
- Cardiovascular Disease Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.
| |
Collapse
|
49
|
Rajakumar P, Anandhan R, Vadla GP, Vellaichamy E. Synthesis and cardio protective biological applications of glucodendrimers by H9C2 cell studies. Carbohydr Polym 2014; 99:403-14. [DOI: 10.1016/j.carbpol.2013.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/12/2013] [Accepted: 08/18/2013] [Indexed: 02/07/2023]
|
50
|
Protective effect of metformin against cisplatin-induced ototoxicity in an auditory cell line. J Assoc Res Otolaryngol 2013; 15:149-58. [PMID: 24297263 DOI: 10.1007/s10162-013-0431-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 11/18/2013] [Indexed: 12/16/2022] Open
Abstract
Metformin, an antidiabetic drug with potent anticancer activity, is known to prevent oxidative stress-induced cell death in several cell types through a mechanism dependent on the mitochondria. In the present study, we investigated the influence of metformin on cisplatin ototoxicity in an auditory cell line. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (Sigma, St. Louis, MO, USA) cell proliferation assay. Oxidative stress and apoptosis were assessed by flow cytometry analysis, Hoechst 33258 staining, reactive oxygen species (ROS) measurement, and western blotting. Intracellular calcium concentration changes were detected using calcium imaging. Pretreatment with 1 mM metformin prior to the application of 20 μM cisplatin significantly decreased the frequency of late apoptosis in HEI-OC1 cells and also significantly attenuated the cisplatin-induced increase in ROS. In addition, metformin inhibited the activation of caspase-3 and levels of poly-ADP-ribose polymerase (PARP). Pretreatment with metformin prevented the cisplatin-induced elevation in intracellular calcium concentrations. We propose that metformin protects against cisplatin-induced ototoxicity by inhibiting the increase in intracellular calcium levels, preventing apoptosis, and limiting ROS production.
Collapse
|