101
|
Markowicz-Piasecka M, Sadkowska A, Huttunen KM, Podsiedlik M, Mikiciuk-Olasik E, Sikora J. An investigation into the pleiotropic activity of metformin. A glimpse of haemostasis. Eur J Pharmacol 2020; 872:172984. [PMID: 32017937 DOI: 10.1016/j.ejphar.2020.172984] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/08/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023]
Abstract
The most characteristic features of type 2 diabetes mellitus (T2DM) are hyperglycaemia and insulin resistance, however, patients with T2DM are at higher risk of cardiovascular disease (CVD) and atherosclerosis. Diabetes, frequently related to metabolic and vascular impairments, is also associated with thrombosis, increased blood coagulation and an imbalance between coagulation and fibrinolysis. Metformin is the most often used oral glucose-lowering agent; its beneficial properties include lowering insulin resistance, weight reduction and cardioprotection. Available data suggest that the advantageous properties of metformin stem from its favourable effects on endothelium, and anti-oxidative and anti-inflammatory properties. This paper reviews the favourable impact of metformin on endothelial function, with particular emphasis on the release of endogenous molecules modulating the state of the vascular endothelium and coagulation. It also summarizes the present knowledge on the influence of metformin on platelet activity and plasma haemostasis, including clot formation, stabilization and fibrinolysis. Its findings confirm that metformin should constitute first line therapy of T2DM subjects; however, more comprehensive methodical studies are required to discover the full potential of this drug.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151, Lodz, Poland.
| | - Adrianna Sadkowska
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151, Lodz, Poland.
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland.
| | - Maria Podsiedlik
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151, Lodz, Poland.
| | - Elżbieta Mikiciuk-Olasik
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151, Lodz, Poland.
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151, Lodz, Poland.
| |
Collapse
|
102
|
Tseng CH. Metformin reduces risk of varicose veins in patients with type 2 diabetes. Diabetes Metab Res Rev 2020; 36:e3206. [PMID: 31322821 DOI: 10.1002/dmrr.3206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 12/28/2022]
Abstract
AIM This population-based retrospective cohort study compared the incidence of varicose veins in an unmatched cohort and a cohort of 1:1 propensity score (PS)-matched pairs of ever and never users of metformin in type 2 diabetes patients. METHODS Patients with new-onset type 2 diabetes during 1999 to 2005 were enrolled from Taiwan's National Health Insurance and followed until December 31, 2011. Analyses were conducted in an unmatched cohort of 123 710 ever users and 15 095 never users and in 15 088 PS-matched pairs of ever users and never users. Hazard ratios were estimated by Cox proportional hazards model incorporated with the inverse probability of treatment weighting using the PS. RESULTS New-onset varicose veins were diagnosed in 126 never users and 633 ever users in the unmatched cohort and in 126 never users and 80 ever users in the matched cohort. The respective incidences were 191.36 and 110.04 per 100 000 person-years in the unmatched cohort and 191.41 and 115.81 per 100 000 person-years in the matched cohort. The hazard ratio for ever versus never users in the unmatched cohort was 0.57 (95% confidence interval, 0.47-0.69) and was 0.60 (0.45-0.80) for the matched cohort. In the unmatched cohort, the hazard ratios for the first, second, and third tertiles of cumulative duration were 1.03 (0.83-1.28), 0.55 (0.44-0.69), and 0.29 (0.23-0.37), respectively. The respective hazard ratios in the matched cohort were 0.97 (0.65-1.43), 0.79 (0.55-1.15), and 0.24 (0.13-0.42). CONCLUSION Metformin use is associated with a lower risk of varicose veins in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
103
|
Yendapally R, Sikazwe D, Kim SS, Ramsinghani S, Fraser‐Spears R, Witte AP, La‐Viola B. A review of phenformin, metformin, and imeglimin. Drug Dev Res 2020; 81:390-401. [DOI: 10.1002/ddr.21636] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/05/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022]
Affiliation(s)
| | - Donald Sikazwe
- Feik School of PharmacyUniversity of the Incarnate Word San Antonio Texas
| | - Subin S. Kim
- Feik School of PharmacyUniversity of the Incarnate Word San Antonio Texas
| | - Sushma Ramsinghani
- Feik School of PharmacyUniversity of the Incarnate Word San Antonio Texas
| | | | - Amy P. Witte
- Feik School of PharmacyUniversity of the Incarnate Word San Antonio Texas
| | - Brittany La‐Viola
- School of PharmacyUniversity of Maryland Eastern Shore Princess Anne Maryland
| |
Collapse
|
104
|
Romualdi D, Versace V, Lanzone A. What is new in the landscape of insulin-sensitizing agents for polycystic ovary syndrome treatment. Ther Adv Reprod Health 2020; 14:2633494120908709. [PMID: 32435760 PMCID: PMC7236839 DOI: 10.1177/2633494120908709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Polycystic ovary syndrome, the most common gynecological endocrinopathy, is burdened with a state of hyperinsulinemia and insulin resistance in 50-80% of affected women. Wherever the origin of these metabolic abnormalities lies, their pathogenetic role in determining, perpetuating, and worsening the clinical traits of the syndrome is ascertained. Many studies have already highlighted possible mechanisms: hyperinsulinemia and insulin resistance may contribute to hyperandrogenemia, chronic anovulation, and other comorbidities of the syndrome by differentially affecting the endocrine glands (ovaries, adrenals, and pituitary) and peripheral tissues (fat mass and skeletal muscle). Based on these evidences, in the past years, thorough research has been focused on the possible role of insulin-sensitizing agents in the treatment of polycystic ovary syndrome. Many compounds were tested to verify their efficacy against polycystic ovary syndrome-related metabolic dysfunction, both relying on previous acquired experiences in the field of diabetes mellitus and experimenting new agents, in particular, those belonging to the class of nutraceuticals. We sought to summarize the most relevant aspects of insulin-sensitizing treatments in polycystic ovary syndrome, by reporting the relevant literature on this topic and by keeping an attentive eye on the newly published international guidelines on polycystic ovary syndrome 2018. This overview encompasses metformin, thiazolidinediones, inositols, alpha-lipoic acid, and GLP1-R analogues. Starting from the analysis of the mechanisms of action, we anchored to the state of the art of the use of these drugs in polycystic ovary syndrome, to the most recent evidences for clinical practice and to the remaining open questions around indications, dose, treatment schedules, and side effects.
Collapse
Affiliation(s)
- Daniela Romualdi
- Department of Woman, Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
- Department Of Woman And Child Health, Division of Obstetrics and Gynecology, Azienda Ospedaliera “Cardinale Panico”, Tricase, Italy
| | - Valeria Versace
- Department of Woman, Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Lanzone
- Department of Woman, Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
105
|
Yang D, Yan J, Deng H, Yang X, Luo S, Zheng X, Lv J, Liang W, Hong M, Wu Z, Yao B, Weng J, Xu W. Effects of Metformin Added to Insulin in Adolescents with Type 1 Diabetes: An Exploratory Crossover Randomized Trial. J Diabetes Res 2020; 2020:7419345. [PMID: 33457425 PMCID: PMC7785393 DOI: 10.1155/2020/7419345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/08/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND To comprehensively assess the effects of metformin added to insulin on metabolic control, insulin sensitivity, and cardiovascular autonomic function in adolescents with type 1 diabetes. MATERIALS AND METHODS This was an exploratory, crossover, randomized trial conducted in adolescents with type 1 diabetes aged 12-18 years old. Participants were randomly received metformin (≤1000 mg/d) added to insulin for 24 weeks followed by insulin monotherapy for a subsequent 24 weeks or vice versa. Blood pressure, body mass index, insulin dose, estimated insulin sensitivity, glycated hemoglobin A1c (HbA1c), and lipid profiles were measured, with a 72-hour continuous glucose monitoring and 24-hour Holter monitoring performed at baseline, 24, and 50 weeks for the assessments of glucose variability and heart rate variability. RESULTS Seventeen patients with mean ± SD age 14.4 ± 2.3 years, body mass index 18.17 ± 1.81 kg/m2, median (IQR) diabetes duration 4.50 (3.58, 6.92) years, and HbA1c 9.0% (8.5%, 9.4%) were enrolled. The between-group difference in HbA1c of 0.28% (95% CI -0.39 to 0.95%) was not significant (P = 0.40). Changes in body mass index, insulin dose, blood pressure, lipid profiles, and estimated insulin sensitivity were similar for metformin add-on vs. insulin monotherapy. Glucose variability also did not differ. Compared with insulin monotherapy, metformin add-on significantly increased multiple heart rate variability parameters. CONCLUSIONS Metformin added to insulin did not improve metabolic control or glucose variability in lean/normal-weight adolescents with type 1 diabetes. However, metformin added to insulin significantly increased heart rate variability, suggesting that metformin might improve cardiovascular autonomic function in this population.
Collapse
Affiliation(s)
- Daizhi Yang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Jinhua Yan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Hongrong Deng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Xubin Yang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Sihui Luo
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences of Medicine, University of Science and Technology of China, Anhui 230026, China
| | - Xueying Zheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences of Medicine, University of Science and Technology of China, Anhui 230026, China
| | - Jing Lv
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Wen Liang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Mengjie Hong
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Department of Cardiovascular Medicine, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Zekai Wu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Bin Yao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences of Medicine, University of Science and Technology of China, Anhui 230026, China
| | - Wen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou 510630, China
| |
Collapse
|
106
|
Markowicz-Piasecka M, Huttunen KM, Sadkowska A, Sikora J. Pleiotropic Activity of Metformin and Its Sulfonamide Derivatives on Vascular and Platelet Haemostasis. Molecules 2019; 25:E125. [PMID: 31905674 PMCID: PMC6982810 DOI: 10.3390/molecules25010125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022] Open
Abstract
As type 2 diabetes mellitus (T2DM) predisposes patients to endothelial cell injury and dysfunction, improvement of vascular function should be an important target for therapy. The aim of this study was to evaluate the effects of metformin, its sulfenamide and sulfonamide derivatives on selected parameters of endothelial and smooth muscle cell function, and platelet activity. Metformin was not found to significantly affect the viability of human umbilical vein endothelial cells (HUVECs) or aortal smooth muscle cells (AoSMC); however, it decreased cell migration by approximately 21.8% in wound healing assays after 24 h stimulation (wound closure 32.5 µm versus 41.5 µm for control). Metformin reduced platelet aggregation manifested by 19.0% decrease in maximum of aggregation (Amax), and 20% reduction in initial platelet aggregation velocity (v0). Furthermore, metformin decreased spontaneous platelet adhesion by 27.7% and ADP-induced adhesion to fibrinogen by 29.6% in comparison to control. Metformin sulfenamide with an n-butyl alkyl chain (compound 1) appeared to exert the most unfavourable effects on AoSMC cell viability (IC50 = 0.902 ± 0.015 μmol/mL), while 4-nitrobenzenesulfonamide (compound 3) and 2-nitrobenzenesulfonamide (compound 4) derivatives of metformin did not affect AoSMC and HUVEC viability at concentrations up to 2.0 μmol/mL. These compounds were also found to significantly reduce migration of smooth muscle cells by approximately 81.0%. Furthermore, sulfonamides 3 and 4 decreased the initial velocity of platelet aggregation by 11.8% and 20.6%, respectively, and ADP-induced platelet adhesion to fibrinogen by 76.3% and 65.6%. Metformin and its p- and o-nitro-benzenesulfonamide derivatives 3, 4 appear to exert beneficial effects on some parameters of vascular and platelet haemostasis.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland;
| | - Adrianna Sadkowska
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| |
Collapse
|
107
|
Larsen AH, Jessen N, Nørrelund H, Tolbod LP, Harms HJ, Feddersen S, Nielsen F, Brøsen K, Hansson NH, Frøkiær J, Poulsen SH, Sörensen J, Wiggers H. A randomised, double‐blind, placebo‐controlled trial of metformin on myocardial efficiency in insulin‐resistant chronic heart failure patients without diabetes. Eur J Heart Fail 2019; 22:1628-1637. [DOI: 10.1002/ejhf.1656] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/04/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Affiliation(s)
| | - Niels Jessen
- Department of Clinical Pharmacology Aarhus University Hospital Aarhus Denmark
- Department of Biomedicine Aarhus University Aarhus Denmark
- Steno Diabetes Centre Aarhus Aarhus University Hospital Aarhus Denmark
| | - Helene Nørrelund
- Department of Clinical Epidemiology, Institute of Clinical Medicine Aarhus University Hospital Aarhus Denmark
| | - Lars Poulsen Tolbod
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
| | | | - Søren Feddersen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | - Flemming Nielsen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | - Kim Brøsen
- Department of Clinical Biochemistry and Pharmacology Odense University Hospital Odense Denmark
| | | | - Jørgen Frøkiær
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
| | | | - Jens Sörensen
- Department of Nuclear Medicine and PET Centre Aarhus University Hospital Aarhus Denmark
- Department of Medical Sciences, Uppsala Clinical Research Centre Uppsala University Uppsala Sweden
| | - Henrik Wiggers
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
108
|
Effects of metformin administration on endocrine-metabolic parameters, visceral adiposity and cardiovascular risk factors in children with obesity and risk markers for metabolic syndrome: A pilot study. PLoS One 2019; 14:e0226303. [PMID: 31821361 PMCID: PMC6903728 DOI: 10.1371/journal.pone.0226303] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
Background Metformin treatment (1000–2000 mg/day) over 6 months in pubertal children and/or adolescents with obesity and hyperinsulinism is associated with a reduction in body mass index (BMI) and the insulin resistance index (HOMA-IR). We aimed to ascertain if long-term treatment (24 months) with lower doses of metformin (850 mg/day) normalizes the endocrine-metabolic abnormalities, improves body composition, and reduces the carotid intima-media thickness (cIMT) in pre-puberal and early pubertal children with obesity. Methods A pilot double-blind, placebo-controlled trial was conducted on 18 pre-puberal and early pubertal (Tanner stage I-II) children with obesity and risk markers for metabolic syndrome. Patients were randomly assigned (1:1) to receive metformin (850 mg/day) or placebo for 24 months. Clinical, biochemical (insulin, lipids, leptin, and high-sensitivity C-reactive protein [hsCRP]), and imaging (body composition [dual-energy X-ray absorptiometry and magnetic resonance imaging]) parameters as well as cIMT (ultrasonography) were assessed at baseline and at 6, 12, and 24 months. Results The 12-month treatment tend to cause a reduction in weight standard deviation scores (SDS), BMI-SDS, leptin, leptin–to–high-molecular-weight (HMW) adiponectin ratio, hsCRP, cIMT, fat mass, and liver fat in metformin-treated children compared with placebo. The effect of metformin on the reduction of BMI-SDS, leptin, leptin-to-HMW adiponectin ratio, hsCRP, and liver fat seemed to be maintained after completing the 24 months of treatment. No changes in insulin sensitivity (HOMA-IR) or adverse effects were detected. Conclusion In this pilot study, metformin treatment in pre-puberal and early pubertal children with obesity seemed to improve body composition and inflammation markers. Our data encourage the development of future fully powered trials using 850 mg/day metformin in young children, highlighting its excellent tolerance and potential long-term benefits.
Collapse
|
109
|
Samuel SM, Varghese E, Kubatka P, Triggle CR, Büsselberg D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules 2019; 9:E846. [PMID: 31835318 PMCID: PMC6995629 DOI: 10.3390/biom9120846] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/25/2022] Open
Abstract
Interest has grown in studying the possible use of well-known anti-diabetic drugs as anti-cancer agents individually or in combination with, frequently used, chemotherapeutic agents and/or radiation, owing to the fact that diabetes heightens the risk, incidence, and rapid progression of cancers, including breast cancer, in an individual. In this regard, metformin (1, 1-dimethylbiguanide), well known as 'Glucophage' among diabetics, was reported to be cancer preventive while also being a potent anti-proliferative and anti-cancer agent. While meta-analysis studies reported a lower risk and incidence of breast cancer among diabetic individuals on a metformin treatment regimen, several in vitro, pre-clinical, and clinical studies reported the efficacy of using metformin individually as an anti-cancer/anti-tumor agent or in combination with chemotherapeutic drugs or radiation in the treatment of different forms of breast cancer. However, unanswered questions remain with regards to areas such as cancer treatment specific therapeutic dosing of metformin, specificity to cancer cells at high concentrations, resistance to metformin therapy, efficacy of combinatory therapeutic approaches, post-therapeutic relapse of the disease, and efficacy in cancer prevention in non-diabetic individuals. In the current article, we discuss the biology of metformin and its molecular mechanism of action, the existing cellular, pre-clinical, and clinical studies that have tested the anti-tumor potential of metformin as a potential anti-cancer/anti-tumor agent in breast cancer therapy, and outline the future prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer drug in the treatment of breast cancer.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
110
|
Abdel Shaheed C, Carland JE, Graham GG, Stocker SL, Smith G, Hicks M, Williams KM, Furlong T, Macdonald P, Greenfield JR, Smith FC, Chowdhury G, Day RO. Is the use of metformin in patients undergoing dialysis hazardous for life? A systematic review of the safety of metformin in patients undergoing dialysis. Br J Clin Pharmacol 2019; 85:2772-2783. [PMID: 31471973 DOI: 10.1111/bcp.14107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/04/2019] [Accepted: 08/18/2019] [Indexed: 12/22/2022] Open
Abstract
AIMS Metformin may have clinical benefits in dialysis patients; however, its safety in this population is unknown. This systematic review evaluated the safety of metformin in dialysis patients. METHODS MEDLINE, Embase, CENTRAL, PsycINFO and the Cochrane Library were searched for randomised controlled trials and observational studies evaluating metformin use in dialysis patients. Three authors reviewed the studies and extracted data. The primary outcomes were mortality, occurrence of lactic acidosis and myocardial infarction (MI) in patients taking metformin during dialysis treatment for ≥12 months (long term). Risk of bias was assessed using Risk Of Bias In Nonrandomised Studies of Interventions (ROBINS-1). Overall quality of evidence was assessed using Grading of Recommendations Assessment, Development and Evaluation (GRADE). RESULTS Fifteen observational studies were eligible; 7 were prospective observational studies and 8 were case reports/case series. No randomised controlled trials were identified. The 7 prospective observational studies (n = 194) reported on cautious metformin use in patients undergoing maintenance dialysis. Only 3 provided long-term follow-up data. In 2 long-term studies of metformin therapy (≤1000 mg/d) in patients undergoing peritoneal dialysis (PD), 1 reported 6 deaths (6/83; 7%) due to major cardiovascular events (3 MI) and the other reported no deaths (0/35). One long-term study of metformin therapy (250 mg to 500 mg thrice weekly) in patients undergoing haemodialysis reported 4 deaths (4/61; 7%) due to major cardiovascular events (2 MI). These findings provide very low-quality evidence as they come from small observational studies. CONCLUSION The evidence regarding the safety of metformin in people undergoing dialysis is inconclusive. Appropriately designed randomised controlled trials are needed to resolve this uncertainty.
Collapse
Affiliation(s)
- Christina Abdel Shaheed
- School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Jane E Carland
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Garry G Graham
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia
| | - Sophie L Stocker
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Greg Smith
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Mark Hicks
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Kenneth M Williams
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia
| | - Timothy Furlong
- Department of Nephrology, St Vincent's Hospital, Sydney, Australia
| | - Peter Macdonald
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Jerry R Greenfield
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia.,Diabetes and Metabolism Division, Garvan Institute of Metabolic Research, Sydney, Australia.,Department of Diabetes and Endocrinology, St Vincent's Hospital, Sydney, Australia
| | - Felicity C Smith
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Gina Chowdhury
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Richard O Day
- Department of Clinical Pharmacology and Toxicology, St Vincent's Hospital, Sydney, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| |
Collapse
|
111
|
Treatment with metformin prevents myocardial ischemia-reperfusion injury via STEAP4 signaling pathway. Anatol J Cardiol 2019; 21:261-271. [PMID: 31062756 PMCID: PMC6528516 DOI: 10.14744/anatoljcardiol.2019.11456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective: The aim of the present study was to investigate the underlying mechanism of metformin in reducing myocardial apoptosis and improving mitochondrial function in rats and H9c2 cells subjected to myocardial ischemia–reperfusion (I/R) or hypoxia–reoxygenation (H/R) injuries, respectively. Methods: Following pretreatment with metformin, male Sprague–Dawley rats were used to establish an I/R model in vivo. Serum creatinine kinase-MB and cardiac troponin T levels were examined by enzyme-linked immunosorbent assay. Infarct size and apoptosis were measured by triphenyl tetrazolium chloride staining and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Pathological changes were evaluated by hematoxylin and eosin staining. H9c2 cells were used to establish an H/R model in vitro. Cell apoptosis and mitochondrial membrane potential (MMP) were examined by flow cytometry and Rhodamine 123. The expression levels of six-transmembrane epithelial antigen of prostate 4 (STEAP4), B-cell lymphoma 2, Bcl-2-associated X protein, and glyceraldehyde 3-phosphate dehydrogenase in both myocardial tissues and H9c2 cells were determined by western blotting. Results: We found that metformin decreased infarct size, increased STEAP4 expression, mitigated myocardial apoptosis, and increased MMP when the models were subjected to H/R or I/R injuries. However, STEAP4 knockdown significantly abrogated the beneficial effect of metformin. Conclusion: We further demonstrated the protective effect of metformin on cardiomyocytes, which might be at least partly attributable to the upregulation of STEAP4. Therefore, STEAP4 might be a new target to decrease apoptosis and rescue mitochondrial function in myocardial I/R injury.
Collapse
|
112
|
Heidari B, Lerman A, Lalia AZ, Lerman LO, Chang AY. Effect of Metformin on Microvascular Endothelial Function in Polycystic Ovary Syndrome. Mayo Clin Proc 2019; 94:2455-2466. [PMID: 31806099 PMCID: PMC8050832 DOI: 10.1016/j.mayocp.2019.06.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 06/07/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the factors that are associated with the effect of metformin on endothelial dysfunction in polycystic ovary syndrome (PCOS). PATIENTS AND METHODS From March 24, 2014, to November 18, 2016, 48 women with PCOS were randomly assigned to 1500 mg/d of metformin (N=29) or no treatment (N=13) for 3 months; 42 patients (29 in the initial treatment group and 13 in the no treatment group) completed the study. Study variables were measured at baseline and after 3 months. Participants who did not receive metformin initially were then treated with metformin for another 3 months, and study variables were measured again. Endothelial function was measured as reactive hyperemia-peripheral arterial tonometry (RH-PAT) from the index finger. RESULTS The age and baseline endothelial function (mean ± SD) of the participants were 32.7±6.9 years and 1.8±0.5, respectively. No notable change was observed in endothelial function after 3 months with metformin compared with no treatment. However, after stratifying participants who received metformin based on baseline endothelial function, there was a significant improvement following metformin treatment in participants with abnormal baseline endothelial function (1.3±0.3 vs 1.7±0.3; P<.001) but not in those with normal baseline endothelial function (2.1±0.4 vs 2.0±0.5; P=.11). CONCLUSION Metformin improves endothelial function in women with PCOS and endothelial dysfunction independent of changes in glucose metabolism, dyslipidemia, or presence of prediabetes. Metformin has a direct effect on endothelial function in PCOS, and measurement of endothelial function can stratify and follow response to metformin treatment in PCOS. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT02086526.
Collapse
Affiliation(s)
- Behnam Heidari
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Antigoni Z Lalia
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Alice Y Chang
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN.
| |
Collapse
|
113
|
Asai A, Shuto Y, Nagao M, Kawahara M, Miyazawa T, Sugihara H, Oikawa S. Metformin Attenuates Early-Stage Atherosclerosis in Mildly Hyperglycemic Oikawa-Nagao Mice. J Atheroscler Thromb 2019; 26:1075-1083. [PMID: 30971640 PMCID: PMC6927811 DOI: 10.5551/jat.48223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: Although metformin treatment has been reported to reduce the risk of cardiovascular events in patients with type 2 diabetes, the underlying mechanisms have not been elucidated fully. Here we assessed atherosclerotic lesion formation in newly established 2 mouse lines with different blood glucose levels (Oikawa-Nagao Diabetes-Prone [ON-DP] and -Resistant [ON-DR]) to evaluate the effect of metformin on early-stage atherosclerosis. Methods: Mildly hyperglycemic ON-DP and normoglycemic ON-DR female mice fed an atherogenic diet for 20 weeks (8–28 weeks of age). During the feeding period, one group of each mouse line received metformin in drinking water (0.1%), while another group received water alone as control. Atherosclerotic lesion formation in the aortic sinus was quantitively analyzed from the oil red O-stained area of the serial sections. Results: Metformin treatment did not affect food intake, body weight, and casual blood glucose levels within each mouse line during the 20-week feeding period. Nevertheless, metformin treatment significantly reduced atherosclerotic lesion formation in the ON-DP mice (59% of control), whereas no significant effect of metformin was observed in the lesion size of the ON-DR mice. Conclusion: Metformin can attenuate early-stage atherogenesis in mildly hyperglycemic ON-DP mice. Pleiotropic effects of metformin, beyond its glucose-lowering action, may contribute to the antiatherogenic property in the early-stage atherosclerosis.
Collapse
Affiliation(s)
- Akira Asai
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School.,Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University
| | - Yuki Shuto
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School
| | - Mototsugu Nagao
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School
| | - Momoyo Kawahara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School
| | - Teruo Miyazawa
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University
| | - Hitoshi Sugihara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School
| | - Shinichi Oikawa
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School
| |
Collapse
|
114
|
Markowicz-Piasecka M, Sikora J, Zajda A, Huttunen KM. Novel halogenated sulfonamide biguanides with anti-coagulation properties. Bioorg Chem 2019; 94:103444. [PMID: 31776031 DOI: 10.1016/j.bioorg.2019.103444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 01/06/2023]
Abstract
Apart from its hypoglycaemic properties, metformin also offers beneficial effects for the cardiovascular system resulting in significant reduction of diabetes-related death, and all-cause mortality. The aim of this study was to synthesize nine new benzenesulfonamide derivatives of metformin with a halogen substituent, and estimate their influence on selected parameters of plasma and vascular hemostasis. The study describes the synthesis of nine benzenesulfonamide biguanides with o-, m-, and p- chloro-, bromo-, and fluoro substituents. All orto- derivatives (chloro- (1), bromo- (4), and fluoro- (7)) significantly prolong prothrombin time (PT) and partially activated thromboplastin time (APTT). In addition compounds 4 and 7 slow the process of fibrin polymerization, and contribute to increased TT. Multiparametric CL-test revealed that compounds 1, 4, 7 and p-fluorobenzenesulfonamide (9) significantly prolong the onset of clot formation, decrease initial clot formation velocity, and maximum clotting. Analysis of human endothelial cell (HUVECs) and human aortal smooth muscle cell (AoSMCs) viability over the entire tested concentration range (0.001-3.0 μmol/mL) indicated that the examined compounds can undergo further tests up to 1.5 µmol/mL concentration without decreasing cellular viability. Furthermore, none of the synthesized compounds exert an unfavourable effect on erythrocyte integrity, and thus do not interact strongly with the lipid-protein bilayer. In summary, chemical modification of the metformin backbone into benzenesulfonamides containing halogen substituents at the o- position leads to the formation of potential agents with stronger anti-coagulant properties than the parent drug, metformin. Therefore, o-halogenated benzenesulfonamides can be regarded as an initial promising step in the development of novel biguanide-based compounds with anti-coagulant properties.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland.
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland.
| | - Agnieszka Zajda
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland.
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland.
| |
Collapse
|
115
|
Tseng C. Metformin Use Is Associated With a Lower Risk of Hospitalization for Heart Failure in Patients With Type 2 Diabetes Mellitus: a Retrospective Cohort Analysis. J Am Heart Assoc 2019; 8:e011640. [PMID: 31630591 PMCID: PMC6898844 DOI: 10.1161/jaha.118.011640] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 09/10/2019] [Indexed: 12/19/2022]
Abstract
Background A beneficial effect of metformin on heart failure requires confirmation. Methods and Results Patients with new-onset type 2 diabetes mellitus during 1999 to 2005 were enrolled from Taiwan's National Health Insurance database and followed up from January 1, 2006, until December 31, 2011. Main analyses were conducted in an unmatched cohort (172 542 metformin ever users and 43 744 never users) and a propensity score matched-pair cohort (matched cohort I, 41 714 ever users and 41 714 never users). Hazard ratios were estimated by Cox hazard regression incorporated with the inverse probability of treatment weighting using the propensity score in the unmatched cohort and by naïve method in the matched cohort I. Results showed that the respective incidence rates of heart failure hospitalization in ever users and never users were 304.25 and 864.31 per 100 000 person-years in the unmatched cohort (hazard ratio, 0.350; 95% CI, 0.329-0.373) and were 469.66 and 817.01 per 100 000 person-years in the matched cohort I (hazard ratio, 0.571; 95% CI, 0.526-0.620). A dose-response pattern was consistently observed while estimating hazard ratios for the tertiles of cumulative duration of metformin therapy. Findings were supported by another propensity score-matched cohort created after excluding 10 potential instrumental variables in the estimation of propensity score (matched cohort II). An approximately 40% lower risk was consistently observed among ever users in different models derived from the matched cohorts I and II, but models from the matched cohort II were less subject to model misspecification. Conclusions Metformin use is associated with a lower risk of heart failure hospitalization.
Collapse
Affiliation(s)
- Chin‐Hsiao Tseng
- Department of Internal MedicineNational Taiwan University College of MedicineTaipeiTaiwan
- Division of Endocrinology and MetabolismDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of Environmental Health and Occupational MedicineNational Health Research InstitutesZhunanTaiwan
| |
Collapse
|
116
|
Ungurianu A, Şeremet O, Gagniuc E, Olaru OT, Guţu C, Grǎdinaru D, Ionescu-Tȋrgovişte C, Marginǎ D, Dǎnciulescu-Miulescu R. Preclinical and clinical results regarding the effects of a plant-based antidiabetic formulation versus well established antidiabetic molecules. Pharmacol Res 2019; 150:104522. [PMID: 31698065 DOI: 10.1016/j.phrs.2019.104522] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/12/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus (DM) is a complex syndrome with debilitating long-term complications, comprising alterations of carbohydrate, protein and lipid metabolisms, along increased oxidative stress and chronic low-grade inflammation. Diet management and plant-based formulations can improve the metabolic status of patients, being used as adjuvants of classic antidiabetic therapy. The purpose of our study was to evaluate the impact of a plant-based antidiabetic formulation (PBAF), containing Vaccinium myrtillus, Ribes nigrum, Rosa canina and Capsicum annuum, on the increased oxidative burden found in diabetes mellitus, comparing it with the effects of metformin and gliclazide. Firstly, we characterized the individual plant-derived components of this formulation and also assessed their in vitro radical scavenging capacity. We devised a preclinical study protocol to examine the impact of the PBAF, along metformin and gliclazide, on tissue histology as well as on the redox status of tissue, mitochondria, serum and serum lipoproteins of alloxan-induced diabetic Wistar rats. Subsequently, we assessed their long-term impact on the redox status of serum and isolated serum lipoproteins of type 2 DM (T2DM) patients, taking into consideration their cardiometabolic profile. In the preclinical stage, we found that PBAF was able to enhance total serum antioxidant defense, while metformin yielded the best results regarding the advanced glycation and protein/lipid oxidation of serum and of serum lipoproteins. The latter also improved overall serum redox status and HDL redox function. Also, antidiabetic treatment seemed to increase mitochondrial redox activity, without overturning overall tissue redox balance. Histologically, liver and brain tissues of treated diabetic rats were fairly similar to those of non-diabetic rats. In T2DM patients, the most striking results involved the effects on serum lipoproteins. The tested PBAF exerted protective antioxidant effects on low-density and, especially, on high density lipoproteins. We conclude that this formulation might constitute a good addition to the well-established pharmacological approach of DM, contributing to the reduction of overall oxidative burden.
Collapse
Affiliation(s)
- Anca Ungurianu
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania
| | - Oana Şeremet
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania
| | - Elvira Gagniuc
- University of Agronomic Sciences and Veterinary Medicine, Faculty of Veterinary Medicine, Splaiul Independenței 105, Bucharest, 050097, Romania
| | - Octavian Tudor Olaru
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania
| | - Claudia Guţu
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania
| | - Daniela Grǎdinaru
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania
| | - Constantin Ionescu-Tȋrgovişte
- "N. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, Ion Movilă 5-7, Bucharest, 030167, Romania
| | - Denisa Marginǎ
- "Carol Davila" University of Medicine and Pharmacy, Faculty of Pharmacy, Traian Vuia 6, Bucharest, 020956, Romania.
| | - Rucsandra Dǎnciulescu-Miulescu
- "N. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, Ion Movilă 5-7, Bucharest, 030167, Romania; "Carol Davila" University of Medicine and Pharmacy, Faculty of Dentistry, Department of Department of Endocrinology, Calea Plevnei 17-23, Bucharest, 020021, Romania
| |
Collapse
|
117
|
Affiliation(s)
- Sanjay Rajagopalan
- Harrington Heart and Vascular Institute, University Hospitals, and The Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Imran Rashid
- Harrington Heart and Vascular Institute, University Hospitals, and The Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
118
|
Sainio A, Takabe P, Oikari S, Salomäki-Myftari H, Koulu M, Söderström M, Pasonen-Seppänen S, Järveläinen H. Metformin decreases hyaluronan synthesis by vascular smooth muscle cells. J Investig Med 2019; 68:383-391. [PMID: 31672719 PMCID: PMC7063400 DOI: 10.1136/jim-2019-001156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2019] [Indexed: 01/09/2023]
Abstract
Metformin is the first-line drug in the treatment of type 2 diabetes worldwide based on its effectiveness and cardiovascular safety. Currently metformin is increasingly used during pregnancy in women with gestational diabetes mellitus, even if the long-term effects of metformin on offspring are not exactly known. We have previously shown that high glucose concentration increases hyaluronan (HA) production of cultured human vascular smooth muscle cells (VSMC) via stimulating the expression of hyaluronan synthase 2 (HAS2). This offers a potential mechanism whereby hyperglycemia leads to vascular macroangiopathy. In this study, we examined whether gestational metformin use affects HA content in the aortic wall of mouse offspring in vivo. We also examined the effect of metformin on HA synthesis by cultured human VSMCs in vitro. We found that gestational metformin use significantly decreased HA content in the intima-media of mouse offspring aortas. In accordance with this, the synthesis of HA by VSMCs was also significantly decreased in response to treatment with metformin. This decrease in HA synthesis was shown to be due to the reduction of both the expression of HAS2 and the amount of HAS substrates, particularly UDP-N-acetylglucosamine. As shown here, gestational metformin use is capable to program reduced HA content in the vascular wall of the offspring strongly supporting the idea, that metformin possesses long-term vasculoprotective effects.
Collapse
Affiliation(s)
- Annele Sainio
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Piia Takabe
- Institute of Biomedicine, University of Eastern Finland-Kuopio Campus, Kuopio, Finland
| | - Sanna Oikari
- Institute of Biomedicine, University of Eastern Finland-Kuopio Campus, Kuopio, Finland.,Institute of Dentistry, University of Eastern Finland-Kuopio Campus, Kuopio, Finland
| | | | - Markku Koulu
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | - Hannu Järveläinen
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Internal Medicine, Satakunta Central Hospital, Pori, Finland
| |
Collapse
|
119
|
Bjornstad P, Schäfer M, Truong U, Cree-Green M, Pyle L, Baumgartner A, Garcia Reyes Y, Maniatis A, Nayak S, Wadwa RP, Browne LP, Reusch JEB, Nadeau KJ. Metformin Improves Insulin Sensitivity and Vascular Health in Youth With Type 1 Diabetes Mellitus. Circulation 2019; 138:2895-2907. [PMID: 30566007 DOI: 10.1161/circulationaha.118.035525] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiovascular disease is the leading cause of mortality in type 1 diabetes mellitus (T1DM) and relates strongly to insulin resistance (IR). Lean and obese adolescents with T1DM have marked IR. Metformin improves surrogate markers of IR in T1DM, but its effect on directly measured IR and vascular health in youth with T1DM is unclear. We hypothesized that adolescents with T1DM have impaired vascular function and that metformin improves this IR and vascular dysfunction. METHODS Adolescents with T1DM and control participants underwent magnetic resonance imaging of the ascending (AA) and descending aorta to assess pulse wave velocity, relative area change, and maximal (WSSMAX) and time-averaged (WSSTA) wall shear stress. Participants with T1DM also underwent assessment of carotid intima-media thickness by ultrasound, brachial distensibility by DynaPulse, fat and lean mass by dual-energy x-ray absorptiometry, fasting laboratories after overnight glycemic control, and insulin sensitivity by hyperinsulinemic-euglycemic clamp (glucose infusion rate/insulin). Adolescents with T1DM were randomized 1:1 to 3 months of 2000 mg metformin or placebo daily, after which baseline measures were repeated. RESULTS Forty-eight adolescents with T1DM who were 12 to 21 years of age (40% body mass index [BMI] ≥90th percentile; 56% female) and 24 nondiabetic control participants of similar age, BMI, and sex distribution were enrolled. Adolescents with T1DM demonstrated impaired aortic health compared with control participants, including elevated AA and descending aorta pulse wave velocity, reduced AA and descending aorta relative area change, and elevated AA and descending aorta WSSMAX and WSSTA. Adolescents with T1DM in the metformin versus placebo group had improved glucose infusion rate/insulin (12.2±3.2 [mg·kg-1·min-1]/μIU/μL versus -2.4±3.6 [mg·kg-1·min-1]/μIU/μL, P=0.005; 18.6±4.8 [mg·lean kg-1·min-1]/μIU/μL versus -3.4±5.6 [mg·lean kg-1·min-1]/μIU/μL, P=0.005) and reduced weight (-0.5±0.5 kg versus 1.6±0.5 kg; P=0.004), BMI (-0.2±0.15 kg/m2 versus 0.4±0.15 kg/m2; P=0.005), and fat mass (-0.7±0.3 kg versus 0.6±0.4 kg; P=0.01). Glucose infusion rate/insulin also improved in normal-weight participants (11.8±4.4 [mg·kg-1·min-1]/μIU/μL versus -4.5±4.4 [mg·kg-1·min-1]/μIU/μL, P=0.02; 17.6±6.7 [mg·lean kg-1·min-1]/μIU/μL versus -7.0±6.7 [mg·lean kg-1·min-1]/μIU/μL, P=0.02). The metformin group had reduced AA WSSMAX (-0.3±0.4 dyne/cm2 versus 1.5±0.5 dyne/cm2; P=0.03), AA pulse wave velocity (-1.1±1.20 m/s versus 4.1±1.6 m/s; P=0.04), and far-wall diastolic carotid intima-media thickness (-0.04±0.01 mm versus -0.00±0.01 mm; P=0.049) versus placebo. CONCLUSIONS Adolescents with T1DM demonstrate IR and impaired vascular health compared with control participants. Metformin improves IR, regardless of baseline BMI, and BMI, weight, fat mass, insulin dose, and aortic and carotid health in adolescents with T1DM. Metformin may hold promise as a cardioprotective intervention in T1DM. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov . Unique identifier: NCT01808690.
Collapse
Affiliation(s)
- Petter Bjornstad
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| | - Michal Schäfer
- Division of Pediatric Cardiology (M.S., U.T.), University of Colorado School of Medicine, Aurora
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora (M.S.)
| | - Uyen Truong
- Division of Pediatric Cardiology (M.S., U.T.), University of Colorado School of Medicine, Aurora
| | - Melanie Cree-Green
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| | - Laura Pyle
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| | - Amy Baumgartner
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| | - Yesenia Garcia Reyes
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| | | | - Sunil Nayak
- Department of Pediatrics (S.N.), University of Colorado School of Medicine, Aurora
- Pediatric Endocrine Associates, Greenwood Village, CO (S.N.)
| | - R Paul Wadwa
- Barbara Davis Center for Diabetes (R.P.W.), University of Colorado School of Medicine, Aurora
| | - Lorna P Browne
- Division of Radiology (L.B.), University of Colorado School of Medicine, Aurora
| | - Jane E B Reusch
- Division of Endocrinology, Rocky Mountain Regional VAMC, Aurora, CO (J.E.B.R.)
| | - Kristen J Nadeau
- Division of Pediatric Endocrinology (P.B., M.C.-G., L.P., A.B., Y.G.R., K.J.N.), University of Colorado School of Medicine, Aurora
| |
Collapse
|
120
|
Morotti E, Giovanni Artini P, Persico N, Battaglia C. Metformin metabolic and vascular effects in overweight/moderately obese hyperinsulinemic PCOS patients treated with contraceptive vaginal ring: a pilot study. Gynecol Endocrinol 2019; 35:854-861. [PMID: 31081406 DOI: 10.1080/09513590.2019.1613361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The aim of this pilot study was to assess how metformin, associated with a contraceptive vaginal ring, may influence lipid and carbohydrate metabolism, fat distribution, and surrogate markers of arterial function. Among 62 patients, 25 were treated with vaginal ring plus metformin and 37 women with only vaginal ring. The effects were assessed after 6 months. The patients were submitted to evaluation of lipid and carbohydrate metabolism; extended view ultrasonographic evaluation of fat distribution; Doppler analysis of ophthalmic artery; brachial artery flow-mediated vasodilatation; oral glucose tolerance test. After 6 months, the body mass index and waist/hip ratio resulted significantly better in patients who associated metformin to vaginal ring. The fasting glucose, insulin, and glucose/insulin ratio, HOMA-IR, glucose, and insulin AUC 120 were significantly improved in metformin group. The ultrasonographic fat analysis resulted significantly better after metformin. The ophthalmic artery PI significantly improved in metformin group. The brachial artery vasodilation was better in metformin treated patients. In conclusion, metformin, associated with vaginal ring, improves the insulin and carbohydrate metabolism, reduces the body weight and android fat distribution. This, associated with the significant improvements of surrogate markers of arterial function, may be responsible of possible cardiovascular and cerebrovascular protective effects.
Collapse
Affiliation(s)
- Elena Morotti
- Department of Obstetrics and Gynecology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | | | - Nicola Persico
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Cesare Battaglia
- Department of Obstetrics and Gynecology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
121
|
|
122
|
Ge CX, Xu MX, Qin YT, Gu TT, Lou DS, Li Q, Hu LF, Wang BC, Tan J. Endoplasmic reticulum stress-induced iRhom2 up-regulation promotes macrophage-regulated cardiac inflammation and lipid deposition in high fat diet (HFD)-challenged mice: Intervention of fisetin and metformin. Free Radic Biol Med 2019; 141:67-83. [PMID: 31153974 DOI: 10.1016/j.freeradbiomed.2019.05.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/04/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
Abstract
Endoplasmic reticulum stress (ERS) has been implicated in obesity-associated cardiac remodeling and dysfunction. Inactive rhomboid protein 2 (iRhom2), also known as Rhbdf2, is an inactive member of the rhomboid intramembrane proteinase family, playing an essential role in regulating inflammation. Nevertheless, the role of ERS-meditated iRhom2 pathway in metabolic stress-induced cardiomyopathy remains unknown. In the study, we showed that 4-PBA, as an essential ERS inhibitor, significantly alleviated high fat diet (HFD)-induced metabolic disorder and cardiac dysfunction in mice. Additionally, lipid deposition in heart tissues was prevented by 4-PBA in HFD-challenged mice. Moreover, 4-PBA blunted the expression of iRhom2, TACE, TNFR2 and phosphorylated NF-κB to prevent HFD-induced expression of inflammatory factors. Further, 4-PBA restrained HFD-triggered oxidative stress by promoting Nrf-2 signaling. Importantly, 4-PBA markedly suppressed cardiac ERS in HFD mice. The anti-inflammation, anti-ERS and anti-oxidant effects of 4-PBA were verified in palmitate (PAL)-incubated macrophages and cardiomyocytes. In addition, promoting ERS could obviously enhance iRhom2 signaling in vitro. Intriguingly, our data demonstrated that PAL-induced iRhom2 up-regulation apparently promoted macrophage to generate inflammatory factors that could promote cardiomyocyte inflammation and lipid accumulation. Finally, interventions by adding fisetin or metformin significantly abrogated metabolic stress-induced cardiomyopathy through the mechanisms mentioned above. In conclusion, this study provided a novel mechanism for metabolic stress-induced cardiomyopathy pathogenesis. Therapeutic strategy to restrain ROS/ERS/iRhom2 signaling pathway could be developed to prevent myocardial inflammation and lipid deposition, consequently alleviating obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Chen-Xu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Min-Xuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China.
| | - Yu-Ting Qin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266100, PR China
| | - Ting-Ting Gu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, PR China
| | - De-Shuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Qiang Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Lin-Feng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Bo-Chu Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China.
| |
Collapse
|
123
|
Abstract
BACKGROUND Metformin (MET) is used as first-line treatment for type 2 diabetes mellitus but has been shown to have pleiotropic effects that have expanded its use to various conditions. Limited current data exist regarding unconventional use within various patient populations. OBJECTIVE The aim of this study was to evaluate US FDA-approved and off-label MET utilization in the US from 2000 to 2015. METHODS We performed a retrospective analysis of outpatient MET prescribing in the US from 2000 to 2015. Data from the Medical Expenditure Panel Survey (MEPS) administered by the Agency of Healthcare Research and Quality were analyzed. Demographic characteristics, including age, sex, socioeconomic status, comorbidities, and region, were analyzed using the MEPS Household Component (HC). Prescription rates were defined as the annual number of MET prescriptions divided by the corresponding population estimate. Population denominators were derived using the MEPS HC. The MEPS estimates US populations based on sampled persons in the target population (civilian, non-institutionalized) for an entire year. MET prescribing is represented by population per 1000 persons. We determined if changes of MET prescribing were uniform across five age groups: < 18 years, 18-29 years, 30-49 years, 50-64 years, and 64 years and older. RESULTS An estimated 553,291,094 MET prescriptions were dispensed in the US from 2000 to 2015. Prescribing rates steadily increased from 2000 to 2015. FDA-approved MET prescription rates increased from 2.27 per 1000 persons in 2000 to 235 per 1000 persons in 2015, while off-label MET prescription rates increased from 0.74 per 1000 persons in 2000 to 20.3 per 1000 persons in 2015. The top indications for off-label MET use were endocrine disorders (45.8%), cardiovascular disorders (18.2%), female reproductive disorders (12.9%), and metabolic disorders (10.9%). MET prescribing rates for FDA-approved indications increased across all age groups in 2000 and 2015, with the most substantial increase seen in adults aged 50-64 years and > 65 years (1.7 per 1000 persons to 20.6 per 1000 persons, and 2.3 per 1000 persons to 18.7 per 1000 persons, respectively). While off-label MET increased across all age groups from 2000 to 2015, a tenfold increase (< 1.0 to 10.6) was seen in adults aged 30-49 years of age. CONCLUSION Overall, MET use has substantially increased within the past 15 years, which was mainly driven by older adults. Our study highlights the emerging prevalence of MET use in both FDA-approved and off-label indications.
Collapse
Affiliation(s)
- Samantha Le
- Division of Pharmacotherapy, University of Texas at Austin College of Pharmacy, UT Health San Antonio, 7703 Floyd Curl Drive MC 6220, San Antonio, TX, 78255, USA
| | - Grace C Lee
- Division of Pharmacotherapy, University of Texas at Austin College of Pharmacy, UT Health San Antonio, 7703 Floyd Curl Drive MC 6220, San Antonio, TX, 78255, USA.
| |
Collapse
|
124
|
Christofides EA. Practical Insights Into Improving Adherence to Metformin Therapy in Patients With Type 2 Diabetes. Clin Diabetes 2019; 37:234-241. [PMID: 31371854 PMCID: PMC6640881 DOI: 10.2337/cd18-0063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
IN BRIEF Adherence to metformin-based treatment regimens for type 2 diabetes is currently suboptimal due to a complex array of patient-, treatment-, and physician-related barriers, including physical and psychological swallowing difficulties associated with large tablets and gastrointestinal disturbances. Patients often avoid discussing these issues with their primary care providers, and delays in addressing them can lead to reduced glycemic control. This article reviews the issues commonly responsible for poor adherence to metformin and presents strategies to improve compliance, including shared decision-making and the use of different metformin formulations, including liquid metformin.
Collapse
|
125
|
Higgins L, Palee S, Chattipakorn SC, Chattipakorn N. Effects of metformin on the heart with ischaemia-reperfusion injury: Evidence of its benefits from in vitro, in vivo and clinical reports. Eur J Pharmacol 2019; 858:172489. [PMID: 31233747 DOI: 10.1016/j.ejphar.2019.172489] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/07/2023]
Abstract
Ischaemia reperfusion (I/R) injury following myocardial infarction reperfusion therapy is a phenomenon that results in further loss of cardiomyocytes and cardiac contractility. Among the potential therapeutics to counter cardiac I/R injury, the antidiabetic drug metformin has shown promising experimental results. This review encompasses evidence available from studies of metformin's protective effects on the heart following cardiac I/R in vitro, ex vivo and in vivo, alongside clinical trials. Experimental data describes potential mechanisms of metformin, including activation of AMPK, an energy sensing kinase with many downstream effects. Suggested effects include upregulation of superoxide dismutases (SODs), which reduce oxidative stress and improve mitochondrial function. Additionally, metformin demonstrates anti-apoptotic effects, most likely by inhibiting mitochondrial permeability transition pore (mPTP) opening, and anti-inflammatory effects, by JNK inhibition. Recent reports of metformin's role in modulating complex I activity of the electron transport chain following cardiac I/R are also presented and discussed. Furthermore, clinical reports present mixed findings, suggesting that beneficial effects may depend on dosage, timing and condition of patients receiving metformin treatment. Conclusively there is an increased need for prospective, placebo-controlled clinical studies to confirm the mechanisms and to demonstrate that metformin is a suitable and safe drug for treatment of cardiac I/R injury.
Collapse
Affiliation(s)
- Louis Higgins
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, England, UK
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
126
|
Bell DSH, Goncalves E. Heart failure in the patient with diabetes: Epidemiology, aetiology, prognosis, therapy and the effect of glucose-lowering medications. Diabetes Obes Metab 2019; 21:1277-1290. [PMID: 30724013 DOI: 10.1111/dom.13652] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 12/24/2022]
Abstract
In people with type 2 diabetes the frequency of heart failure (HF) is increased and mortality from HF is higher than with non-diabetic HF. The increased frequency of HF is attributable to the cardiotoxic tetrad of ischaemic heart disease, left ventricular hypertrophy, diabetic cardiomyopathy and an extracellular volume expansion resistant to atrial natriuretic peptides. Activation of the renin-angiotensin-aldosterone system and sympathetic nervous systems results in cardiac remodelling, which worsens cardiac function. Reversal of remodelling can be achieved, and cardiac function improved in people with HF with reduced ejection fraction (HFrEF) by treatment with angiotensin-converting enzyme inhibitors and β-blockers. However, with HF with preserved ejection fraction (HFpEF), only therapy for the underlying risk factors helps. Blockers of mineralocorticoid receptors may be beneficial in both HFrEF and HFpEF. Glucose-lowering drugs can have a negative effect (insulin, sulphonylureas, dipeptidyl peptidase-4 inhibitors and thiazolidinediones), a neutral effect (α-glucosidase inhibitors and glucagon-like peptide-1 receptor agonists) or a positive effect (sodium-glucose co-transporter-2 inhibitors and metformin).
Collapse
|
127
|
Wang X, Luo C, Mao XY, Li X, Yin JY, Zhang W, Zhou HH, Liu ZQ. Metformin reverses the schizophrenia-like behaviors induced by MK-801 in rats. Brain Res 2019; 1719:30-39. [PMID: 31121159 DOI: 10.1016/j.brainres.2019.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Schizophrenia is known to be a complex and disabling psychiatric disorder. Dopamine receptor antagonists have a significant therapeutic effect in improving the positive symptoms that are associated with the illness. Therefore, dopamine receptor antagonists are commonly used in the treatment of schizophrenia; however, they do not achieve satisfactory results in improving negative symptoms and cognitive impairment. Metformin, widely known as an antidiabetic drug, has been found to enhance spatial memory formation and improve anxiety-like behaviors in rodents. Metformin's neuroprotective effect has been well documented in several neurological disorders including Alzheimer's disease, Parkinson's disease, strokes, Huntington's disease, and seizures. In the present study, we used a rat model to explore the effect of metformin on schizophrenia-like behaviors induced by MK-801 (dizocilpine), an N-methyl-D-aspartate (NMDA) receptor antagonist. We found that the pre-pulse inhibition (PPI) deficit caused by MK-801 could be alleviated by metformin. The hyperlocomotion in the open field test induced by chronic treatment of MK-801 was reversed by administration of metformin. Metformin has no effect on the baseline level of anxiety in normal naive rats, while metformin could relieve the anxiety-like behaviors in MK-801-treatment rats, though this effect is not reaching a significant level. Additionally, metformin could significantly ameliorate working memory impairments induced by MK-801. Moreover, the increased level of phosphorylation of Akt and GSK3β in the frontal cortex induced by MK-801 was normalized by metformin. In conclusion, our results demonstrate that metformin improved schizophrenia-like symptoms in rats, and is therefore a potential agent for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Xu Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Chao Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; School of Life Sciences, Central South University, Changsha, Hunan 410078, PR China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
128
|
Yoshida GJ, Azuma A, Miura Y, Orimo A. Activated Fibroblast Program Orchestrates Tumor Initiation and Progression; Molecular Mechanisms and the Associated Therapeutic Strategies. Int J Mol Sci 2019; 20:ijms20092256. [PMID: 31067787 PMCID: PMC6539414 DOI: 10.3390/ijms20092256] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Neoplastic epithelial cells coexist in carcinomas with various non-neoplastic stromal cells, together creating the tumor microenvironment. There is a growing interest in the cross-talk between tumor cells and stromal fibroblasts referred to as carcinoma-associated fibroblasts (CAFs), which are frequently present in human carcinomas. CAF populations extracted from different human carcinomas have been shown to possess the ability to influence the hallmarks of cancer. Indeed, several mechanisms underlying CAF-promoted tumorigenesis are elucidated. Activated fibroblasts in CAFs are characterized as alpha-smooth muscle actin-positive myofibroblasts and actin-negative fibroblasts, both of which are competent to support tumor growth and progression. There are, however, heterogeneous CAF populations presumably due to the diverse sources of their progenitors in the tumor-associated stroma. Thus, molecular markers allowing identification of bona fide CAF populations with tumor-promoting traits remain under investigation. CAFs and myofibroblasts in wound healing and fibrosis share biological properties and support epithelial cell growth, not only by remodeling the extracellular matrix, but also by producing numerous growth factors and inflammatory cytokines. Notably, accumulating evidence strongly suggests that anti-fibrosis agents suppress tumor development and progression. In this review, we highlight important tumor-promoting roles of CAFs based on their analogies with wound-derived myofibroblasts and discuss the potential therapeutic strategy targeting CAFs.
Collapse
Affiliation(s)
- Go J Yoshida
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Arata Azuma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 1138603, Japan.
| | - Yukiko Miura
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 1138603, Japan.
| | - Akira Orimo
- Department of Molecular Pathogenesis, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
129
|
Markowicz-Piasecka M, Huttunen KM, Broncel M, Sikora J. Sulfenamide and Sulfonamide Derivatives of Metformin - A New Option to Improve Endothelial Function and Plasma Haemostasis. Sci Rep 2019; 9:6573. [PMID: 31024058 PMCID: PMC6484023 DOI: 10.1038/s41598-019-43083-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multi-factorial disease which can cause multiple organ dysfunction, including that of the vascular endothelium. The aim of the present study was to evaluate the effects of metformin, and its sulfenamide and sulfonamide derivatives (compounds 1–8) on the selected markers of endothelial function and blood coagulation. The integrity of endothelial cells(ECs) was examined using the real-time cell electric impedance system. Tissue Factor(TF) production, the release of von Willebrand Factor (vWF) and tissue plasminogen activator(t-PA) from ECs were determined using immunoenzymatic assays, while the process of platelet thrombus formation using the Total Thrombus-Formation Analysis System. Sulfenamide with n-butyl alkyl chain(3) does not interfere with ECs integrity, and viability (nCI(24h) = 1.03 ± 0.03 vs. 1.06 ± 0.11 for control), but possesses anticoagulation properties manifested by prolonged platelet-dependent thrombus formation (Occlusion Time 370.3 ± 77.0 s vs. 286.7 ± 65.5 s for control) in semi-physiological conditions. Both p- and o-nitro-benzenesulfonamides (compounds7,8) exhibit anti-coagulant properties demonstrated by decreased vWF release and prolonged parameters of platelet thrombus formation and total blood thrombogenicity. In conclusion, chemical modification of metformin scaffold into sulfenamides or sulfonamides might be regarded as a good starting point for the design and synthesis of novel biguanide-based compounds with anticoagulant properties and valuable features regarding endothelial function.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland.
| | - Kristiina M Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211, Kuopio, Finland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151, Lodz, Poland
| |
Collapse
|
130
|
Tseng CH. Metformin and risk of chronic obstructive pulmonary disease in diabetes patients. DIABETES & METABOLISM 2019; 45:184-190. [PMID: 29804817 DOI: 10.1016/j.diabet.2018.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/28/2018] [Accepted: 05/06/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE This study aimed to investigate whether metformin can affect risk of chronic obstructive pulmonary disease (COPD) in type 2 diabetes (T2D) patients. METHODS T2D patients newly diagnosed during 1999-2005 were enrolled from the reimbursement database of Taiwan's National Health Insurance system and followed up to 31 December 2011. Analyses were conducted in an unmatched cohort (92,272 ever-users and 10,697 never-users of metformin) and a propensity score (PS) matched pair cohort (10,697 ever-users and 10,697 never-users). Cox regression incorporated into the inverse probability of treatment weighting using the PS was used to estimate hazard ratios (HRs). RESULTS In the unmatched cohort, 2573 never-users and 13,840 ever-users developed COPD with respective incidences of 5994.64 and 3393.19 per 100,000 person-years. The overall HR was 0.560 (95% confidence interval [CI]: 0.537-0.584). HRs for the first (<25.27months), second (25.27-55.97months) and third (>55.97months) tertiles of cumulative duration were 1.021 (0.975-1.070), 0.575 (0.548-0.603) and 0.265 (0.252-0.280), respectively. Analyses of the matched cohort showed an overall HR of 0.643 (0.605-0.682), with HRs of 1.212 (1.122-1.309), 0.631 (0.578-0.689) and 0.305 (0.273-0.340) for the respective tertiles. CONCLUSION A reduced risk of COPD is observed in metformin users with T2D.
Collapse
Affiliation(s)
- C-H Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan, Republic of China; Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China; Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan, Republic of China.
| |
Collapse
|
131
|
Yaribeygi H, Butler AE, Barreto GE, Sahebkar A. Antioxidative potential of antidiabetic agents: A possible protective mechanism against vascular complications in diabetic patients. J Cell Physiol 2019; 234:2436-2446. [DOI: 10.1002/jcp.27278] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/26/2018] [Indexed: 08/30/2023]
Abstract
AbstractMany vascular complications are related to exposure of tissues to elevated levels of glucose, a condition that promotes oxidative stress. The primary goal of antidiabetic medication is for normalization of blood glucose. However, antidiabetic medications may have antioxidant effects that go beyond their hypoglycemic influences. Therefore, antidiabetic drugs may be doubly beneficial in preventing diabetic complications. Vascular dysfunction due to uncontrolled diabetes is a serious complication of the disease and one which has a severe impact on quality of life. Readjustment of the oxidative balance in subjects with diabetes, and the positive effects thereof is a topic of intense interest at present. In the current review, we highlight the antioxidant effects of antidiabetic medications which may prevent or delay the onset of vascular dysfunction.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | | | - George E. Barreto
- Departamento de Nutrición y Bioquímica Facultad de Ciencias Pontificia Universidad Javeriana Bogotá D.C. Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile Santiago Chile
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
132
|
Elksnis A, Martinell M, Eriksson O, Espes D. Heterogeneity of Metabolic Defects in Type 2 Diabetes and Its Relation to Reactive Oxygen Species and Alterations in Beta-Cell Mass. Front Physiol 2019; 10:107. [PMID: 30837889 PMCID: PMC6383038 DOI: 10.3389/fphys.2019.00107] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex and heterogeneous disease which affects millions of people worldwide. The classification of diabetes is at an interesting turning point and there have been several recent reports on sub-classification of T2D based on phenotypical and metabolic characteristics. An important, and perhaps so far underestimated, factor in the pathophysiology of T2D is the role of oxidative stress and reactive oxygen species (ROS). There are multiple pathways for excessive ROS formation in T2D and in addition, beta-cells have an inherent deficit in the capacity to cope with oxidative stress. ROS formation could be causal, but also contribute to a large number of the metabolic defects in T2D, including beta-cell dysfunction and loss. Currently, our knowledge on beta-cell mass is limited to autopsy studies and based on comparisons with healthy controls. The combined evidence suggests that beta-cell mass is unaltered at onset of T2D but that it declines progressively. In order to better understand the pathophysiology of T2D, to identify and evaluate novel treatments, there is a need for in vivo techniques able to quantify beta-cell mass. Positron emission tomography holds great potential for this purpose and can in addition map metabolic defects, including ROS activity, in specific tissue compartments. In this review, we highlight the different phenotypical features of T2D and how metabolic defects impact oxidative stress and ROS formation. In addition, we review the literature on alterations of beta-cell mass in T2D and discuss potential techniques to assess beta-cell mass and metabolic defects in vivo.
Collapse
Affiliation(s)
- Andris Elksnis
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Mats Martinell
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
133
|
Natali A, Nesti L, Venturi E, Shore AC, Khan F, Gooding K, Gates PE, Looker HC, Dove F, Goncalves I, Persson M, Nilsson J. Metformin is the key factor in elevated plasma growth differentiation factor-15 levels in type 2 diabetes: A nested, case-control study. Diabetes Obes Metab 2019; 21:412-416. [PMID: 30178545 DOI: 10.1111/dom.13519] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023]
Abstract
Produced as a tissue defence response to hypoxia and inflammation, growth differentiation factor-15 (GDF-15) is elevated in people receiving metformin treatment. To gain insight into the relationship of GDF-15 with metformin and major cardiovascular risk factors, we analysed the data from the SUMMIT cohort (n = 1438), a four-centre, nested, case-control study aimed at verifying whether biomarkers of atherosclerosis differ according to the presence of type 2 diabetes and cardiovascular disease. While in univariate analysis, major cardiovascular risk factors, with the exception of gender and cholesterol, increased similarly and linearly across GDF-15 quartiles, the independent variables associated with GDF-15, both in participants with and without diabetes, were age, plasma creatinine, N-terminal pro-brain natriuretic peptide, diuretic use, smoking exposure and glycated haemoglobin. In participants with diabetes, metformin treatment was associated with a 40% rise in GDF-15 level, which was independent of the other major factors, and largely explained their elevated GDF-15 levels. The relatively high GDF-15 bioavailability might partly explain the protective cardiovascular effects of metformin.
Collapse
Affiliation(s)
- Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lorenzo Nesti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Venturi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Angela C Shore
- Diabetes and Vascular Medicine Research Centre, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Faisel Khan
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Kim Gooding
- Diabetes and Vascular Medicine Research Centre, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Phillip E Gates
- Diabetes and Vascular Medicine Research Centre, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Helen C Looker
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Fiona Dove
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
134
|
Chin-Hsiao T. Metformin and the Risk of Dementia in Type 2 Diabetes Patients. Aging Dis 2019; 10:37-48. [PMID: 30705766 PMCID: PMC6345339 DOI: 10.14336/ad.2017.1202] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/02/2017] [Indexed: 12/26/2022] Open
Abstract
This retrospective cohort study investigated dementia risk associated with metformin use in type 2 diabetes patients by using the reimbursement database of the Taiwan's National Health Insurance. The patients had new-onset diabetes during 1999-2005 and were followed up until December 31, 2011. An unmatched cohort of 147,729 ever users and 15,676 never users of metformin were identified, and a matched-pair cohort of 15,676 ever users and 15,676 never users was created by propensity score (PS). Hazard ratios were estimated by Cox regression incorporated with the inverse probability of treatment weighting using PS. Results showed that in the unmatched cohort, 713 never users and 3943 ever users developed dementia with respective incidence of 1029.20 and 570.03 per 100,000 person-years. The overall hazard ratio was 0.550 (95% confidence interval: 0.508-0.596). The hazard ratio for the first (<27.0 months), second (27.0-58.1 months) and third (>58.1 months) tertile of cumulative duration of metformin therapy was 0.975 (0.893-1.066), 0.554 (0.506-0.607) and 0.286 (0.259-0.315), respectively. Analyses in the matched cohort showed an overall hazard ratio of 0.707 (0.632-0.791) and the hazard ratio for the respective tertile was 1.279 (1.100-1.488), 0.704 (0.598-0.829) and 0.387 (0.320-0.468). In conclusion, metformin use is associated with a reduced dementia risk.
Collapse
Affiliation(s)
- Tseng Chin-Hsiao
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
135
|
Li X, Liu J, Lu Q, Ren D, Sun X, Rousselle T, Tan Y, Li J. AMPK: a therapeutic target of heart failure-not only metabolism regulation. Biosci Rep 2019; 39:BSR20181767. [PMID: 30514824 PMCID: PMC6328861 DOI: 10.1042/bsr20181767] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/19/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is a serious disease with high mortality. The incidence of this disease has continued to increase over the past decade. All cardiovascular diseases causing dysfunction of various physiological processes can result in HF. AMP-activated protein kinase (AMPK), an energy sensor, has pleiotropic cardioprotective effects and plays a critical role in the progression of HF. In this review, we highlight that AMPK can not only improve the energy supply in the failing heart by promoting ATP production, but can also regulate several important physiological processes to restore heart function. In addition, we discuss some aspects of some potential clinical drugs which have effects on AMPK activation and may have value in treating HF. More studies, especially clinical trials, should be done to evaluate manipulation of AMPK activation as a potential means of treating HF.
Collapse
Affiliation(s)
- Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Jia Liu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| | - Qingguo Lu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
- Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, 37 Guoxue Lane, Chengdu 610041, China
| | - Di Ren
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Xiaodong Sun
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Thomas Rousselle
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Yi Tan
- Pediatic Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, U.S.A
- Wendy L. Novak Diabetes Care Center, University of Louisville, Louisville, KY, U.S.A
| | - Ji Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A.
| |
Collapse
|
136
|
Yadav SK, Kambis TN, Mishra PK. Regulating Inflammatory Cytokines in the Diabetic Heart. OXIDATIVE STRESS IN HEART DISEASES 2019:427-436. [DOI: 10.1007/978-981-13-8273-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
137
|
Mabuza LP, Gamede MW, Maikoo S, Booysen IN, Ngubane PS, Khathi A. Cardioprotective effects of a ruthenium (II) Schiff base complex in diet-induced prediabetic rats. Diabetes Metab Syndr Obes 2019; 12:217-223. [PMID: 30858714 PMCID: PMC6385740 DOI: 10.2147/dmso.s183811] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prediabetes and the onset of cardiovascular diseases (CVD) are strongly related. Prolonged hyperglycemia has been identified as a major contributing factor in the pathogenesis of CVD and diabetic complications. The management of hyperglycemia and prediabetes-associated vascular complications rely on pharmacotherapy and lifestyle intervention strategies. However, patients still take the conventional drugs and neglect lifestyle intervention; therefore, newer alternative drugs are required. The synthesized ruthenium Schiff base complex has been shown to have elevated biological and antidiabetic activity. Thus, the research investigated the cardio-protective effects of ruthenium (II) Schiff base complex in diet-induced prediabetic (PD) rats. MATERIALS AND METHODS The rats were randomly allocated to respective groups and treated for 12 weeks. Ruthenium (15 mg/kg) was administered to PD rats once a day every third day. Blood pressure and plasma glucose were monitored throughout the study. Blood and heart tissue were collected for biochemical assays. RESULTS Ruthenium complex with dietary intervention lead to reduced mean arterial blood pressure which correlated with a restored heart to body weight ratio. Additionally, there was a significant decrease in tissue malondialdehyde and increased superoxide dismutase and glutathione peroxidase concentration in both the plasma and heart tissue. Furthermore, there was a decrease in plasma triglycerides, low-density lipoprotein with an increased high-density lipoprotein concentration in ruthenium-treated rats. This was further evidenced by reduced plasma tumor necrosis factor-α, IL-6, and cardiac C-reactive protein concentrations in ruthenium-treated rats. CONCLUSION Ruthenium coupled with dietary intervention decreased the risk of developing cardiac injury, thus preventing CVD in prediabetes. Therefore, this complex may be a beneficial therapeutic agent in the prevention of PD cardiovascular complications.
Collapse
Affiliation(s)
- Lindokuhle Patience Mabuza
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa,
| | - Mlindeli Wilkinson Gamede
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa,
| | - Sanam Maikoo
- Department of Chemistry, School of Chemistry and Physics, College of Agricultural, Engineering and Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Irvin Noel Booysen
- Department of Chemistry, School of Chemistry and Physics, College of Agricultural, Engineering and Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Phikelelani Siphosethu Ngubane
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa,
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa,
| |
Collapse
|
138
|
Corremans R, Vervaet BA, D'Haese PC, Neven E, Verhulst A. Metformin: A Candidate Drug for Renal Diseases. Int J Mol Sci 2018; 20:E42. [PMID: 30583483 PMCID: PMC6337137 DOI: 10.3390/ijms20010042] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Over the past decades metformin has been the optimal first-line treatment for type 2 diabetes mellitus (T2DM). Only in the last few years, it has become increasingly clear that metformin exerts benign pleiotropic actions beyond its prescribed use and ongoing investigations focus on a putative beneficial impact of metformin on the kidney. Both acute kidney injury (AKI) and chronic kidney disease (CKD), two major renal health issues, often result in the need for renal replacement therapy (dialysis or transplantation) with a high socio-economic impact for the patients. Unfortunately, to date, effective treatment directly targeting the kidney is lacking. Metformin has been shown to exert beneficial effects on the kidney in various clinical trials and experimental studies performed in divergent rodent models representing different types of renal diseases going from AKI to CKD. Despite growing evidence on metformin as a candidate drug for renal diseases, in-depth research is imperative to unravel the molecular signaling pathways responsible for metformin's renoprotective actions. This review will discuss the current state-of-the-art literature on clinical and preclinical data, and put forward potential cellular mechanisms and molecular pathways by which metformin ameliorates AKI/CKD.
Collapse
Affiliation(s)
- Raphaëlle Corremans
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Benjamin A Vervaet
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium.
| |
Collapse
|
139
|
O’Brien MJ, Karam SL, Wallia A, Kang RH, Cooper AJ, Lancki N, Moran MR, Liss DT, Prospect TA, Ackermann RT. Association of Second-line Antidiabetic Medications With Cardiovascular Events Among Insured Adults With Type 2 Diabetes. JAMA Netw Open 2018; 1:e186125. [PMID: 30646315 PMCID: PMC6324353 DOI: 10.1001/jamanetworkopen.2018.6125] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022] Open
Abstract
Importance Understanding cardiovascular outcomes of initiating second-line antidiabetic medications (ADMs) may help inform treatment decisions after metformin alone is not sufficient or not tolerated. To date, no studies have compared the cardiovascular effects of all major second-line ADMs during this early decision point in the pharmacologic management of type 2 diabetes. Objective To examine the association of second-line ADM classes with major adverse cardiovascular events. Design, Setting, and Participants Retrospective cohort study among 132 737 insured adults with type 2 diabetes who started therapy with a second-line ADM after taking either metformin alone or no prior ADM. This study used 2011-2015 US nationwide administrative claims data. Data analysis was performed from January 2017 to October 2018. Exposures Dipeptidyl peptidase 4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, sodium-glucose cotransporter 2 (SGLT-2) inhibitors, thiazolidinediones (TZDs), basal insulin, and sulfonylureas or meglitinides (both referred to as sulfonylureas hereafter). The DPP-4 inhibitors served as the comparison group in all analyses. Main Outcomes and Measures The primary outcome was time to first cardiovascular event after starting the second-line ADM. This composite outcome was based on hospitalization for the following cardiovascular conditions: congestive heart failure, stroke, ischemic heart disease, or peripheral artery disease. Results Among 132 737 insured adult patients with type 2 diabetes (men, 55%; aged 45-64 years, 58%; white, 63%), there were 3480 incident cardiovascular events during 169 384 person-years of follow-up. Patients were censored after the first cardiovascular event, discontinuation of insurance coverage, transition from International Classification of Diseases, Ninth Revision (ICD-9) to end of ICD-9 coding, or 2 years of follow-up. After adjusting for patient, prescriber, and health plan characteristics, the risk of composite cardiovascular events after starting GLP-1 receptor agonists was lower than DPP-4 inhibitors (hazard ratio [HR], 0.78; 95% CI, 0.63-0.96), but this finding was not significant in all sensitivity analyses. Cardiovascular event rates after starting treatment with SGLT-2 inhibitors (HR, 0.81; 95% CI, 0.57-1.53) and TZDs (HR, 0.92; 95% CI, 0.76-1.11) were not statistically different from DPP-4 inhibitors. The comparative risk of cardiovascular events was higher after starting treatment with sulfonylureas (HR, 1.36; 95% CI, 1.23-1.49) or basal insulin (HR, 2.03; 95% CI, 1.81-2.27) than DPP-4 inhibitors. Conclusions and Relevance Among insured adult patients with type 2 diabetes initiating second-line ADM therapy, the short-term cardiovascular outcomes of GLP-1 receptor agonists, SGLT-2 inhibitors, and DPP-4 inhibitors were similar. Higher cardiovascular risk was associated with use of sulfonylureas or basal insulin compared with newer ADM classes. Clinicians may consider prescribing GLP-1 receptor agonists, SGLT-2 inhibitors, or DPP-4 inhibitors more routinely after metformin rather than sulfonylureas or basal insulin.
Collapse
Affiliation(s)
- Matthew J. O’Brien
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Susan L. Karam
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amisha Wallia
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Raymond H. Kang
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Andrew J. Cooper
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Nicola Lancki
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Margaret R. Moran
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Now with Oak Street Health, Chicago, Illinois
| | - David T. Liss
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Ronald T. Ackermann
- Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
140
|
Kalafat E, Sukur YE, Abdi A, Thilaganathan B, Khalil A. Metformin for prevention of hypertensive disorders of pregnancy in women with gestational diabetes or obesity: systematic review and meta-analysis of randomized trials. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2018; 52:706-714. [PMID: 29749110 DOI: 10.1002/uog.19084] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Metformin has been reported to reduce the risk of pre-eclampsia. It is also known to influence soluble fms-like tyrosine kinase-1 level, which correlates significantly with the gestational age at onset and severity of pre-eclampsia. The main aim of this systematic review and meta-analysis of randomized trials was to determine whether metformin use is associated with the incidence of hypertensive disorders of pregnancy (HDP). METHODS MEDLINE (1947 to September 2017), Scopus (1970 to September 2017) and the Cochrane Library (inception to September 2017) were searched for relevant citations in the English language. Only randomized controlled trials on metformin use, reporting the incidence of pre-eclampsia or pregnancy-induced hypertension, were included. Studies on populations with a high probability of metformin use prior to randomization (those with type II diabetes or polycystic ovary syndrome) were excluded. Random-effects models with the Mantel-Haenszel method were used for subgroup analyses. Bayesian random-effects meta-regression was used to summarize the evidence. RESULTS In total, 3337 citations matched the search criteria. After evaluating 2536 abstracts and performing full-text review of 52 studies, 15 were included in the review. In women with gestational diabetes, metformin use was associated with a reduced risk of pregnancy-induced hypertension when compared with insulin (relative risk (RR), 0.56; 95% CI, 0.37-0.85; I2 = 0%; 1260 women) and a non-significantly reduced risk of pre-eclampsia (RR, 0.83; 95% CI, 0.60-1.14; I2 = 0%; 1724 women). In obese women, when compared with placebo, metformin use was associated with a non-significant reduction in risk of pre-eclampsia (RR, 0.74; 95% CI, 0.09-6.28; I2 = 86%; 840 women). In women with gestational diabetes, metformin use was also associated with a non-significant reduction in risk of any HDP (RR, 0.71; 95% CI, 0.41-1.25; I2 = 0%; 556 women) when compared with glyburide. When studies were combined using Bayesian random-effects meta-regression, with treatment type as a covariate, the posterior probabilities of metformin having a beneficial effect on the prevention of pre-eclampsia, pregnancy-induced hypertension and any HDP were 92.7%, 92.8% and 99.2%, respectively, when compared with any other treatment or placebo. CONCLUSIONS There is a high probability that metformin use is associated with reduced HDP incidence when compared with other treatments or placebo. The small number of studies included in the analysis, the low quality of evidence and the clinical heterogeneity preclude generalization of these results to broader populations. Given the clinical importance of this topic and the magnitude of effect observed in this meta-analysis, further prospective trials are urgently needed. Copyright © 2018 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- E Kalafat
- Ankara University Faculty of Medicine, Department of Obstetrics and Gynecology, Ankara, Turkey
- Middle East Technical University, Department of Statistics, Ankara, Turkey
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
| | - Y E Sukur
- Ankara University Faculty of Medicine, Department of Obstetrics and Gynecology, Ankara, Turkey
| | - A Abdi
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
| | - B Thilaganathan
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - A Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
141
|
Chronic Intracerebroventricular Infusion of Metformin Inhibits Salt-Sensitive Hypertension via Attenuation of Oxidative Stress and Neurohormonal Excitation in Rat Paraventricular Nucleus. Neurosci Bull 2018; 35:57-66. [PMID: 30426340 DOI: 10.1007/s12264-018-0308-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/26/2018] [Indexed: 01/15/2023] Open
Abstract
Metformin (MET), an antidiabetic agent, also has antioxidative effects in metabolic-related hypertension. This study was designed to determine whether MET has anti-hypertensive effects in salt-sensitive hypertensive rats by inhibiting oxidative stress in the hypothalamic paraventricular nucleus (PVN). Salt-sensitive rats received a high-salt (HS) diet to induce hypertension, or a normal-salt (NS) diet as control. At the same time, they received intracerebroventricular (ICV) infusion of MET or vehicle for 6 weeks. We found that HS rats had higher oxidative stress levels and mean arterial pressure (MAP) than NS rats. ICV infusion of MET attenuated MAP and reduced plasma norepinephrine levels in HS rats. It also decreased reactive oxygen species and the expression of subunits of NAD(P)H oxidase, improved the superoxide dismutase activity, reduced components of the renin-angiotensin system, and altered neurotransmitters in the PVN. Our findings suggest that central MET administration lowers MAP in salt-sensitive hypertension via attenuating oxidative stress, inhibiting the renin-angiotensin system, and restoring the balance between excitatory and inhibitory neurotransmitters in the PVN.
Collapse
|
142
|
Favorable outcomes of metformin on coronary microvasculature in experimental diabetic cardiomyopathy. J Mol Histol 2018; 49:639-649. [PMID: 30317407 DOI: 10.1007/s10735-018-9801-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023]
Abstract
Although metformin is widely prescribed in diabetes, its use with associated cardiac dysfunction remains debatable. In the current study, we investigated the effect of metformin on coronary microvasculature in experimental diabetic cardiomyopathy (DCM) induced by streptozotocin. Administration of metformin after induction of DCM, reversed almost all cardiomyocyte degenerative changes induced by DCM. Metformin diminished the significantly increased (p < 0.05) collagen deposited in the DCM. In addition metformin had improved the density of the significantly decreased arteriolar (αSMA+) and capillary (CD31+) coronary microvasculature compared to that of the DCM and non-diabetics (ND) with downregulation of the significantly increased expression (p < 0.05) of COL-I, III, TGF-β, CTGF, ICAM and VCAM genes. Therefore metformin may be beneficial in limiting the fibrotic and the vascular remodeling occurring in DCM at the genetic as well as the structural levels.
Collapse
|
143
|
Hua J, Liu Z, Liu Z, An D, Lai W, Zhan Q, Zeng Q, Ren H, Xu D. Metformin Increases Cardiac Rupture After Myocardial Infarction via the AMPK-MTOR/PGC-1α Signaling Pathway in Rats with Acute Myocardial Infarction. Med Sci Monit 2018; 24:6989-7000. [PMID: 30275441 PMCID: PMC6180847 DOI: 10.12659/msm.910930] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Cardiac rupture often occurs after acute myocardial infarction due to complex and unclear pathogenesis. This study investigated whether metformin increases the incidence of cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway. Material/Methods An acute myocardial infarction (MI) mouse model was established. A series of experiments involving RT-qPCR, Western blot, TUNEL staining, and Masson staining were performed in this study. Results Myocardial infarction occurred, resulting in the cardiac rupture, and the expression level of PGC-1α increased in the cardiac myocardium. Meanwhile, the proportion of myocardial NT-PGC-1α/PGC-1α decreased. The expression level of myocardial PGC-1α in MI mice with cardiac rupture after MI was significantly higher than that in the mice without cardiac rupture, and the ratio of myocardial NT-PGC-1α/PGC-1α was low. In addition, increasing the dose of metformin significantly increased the incidence of cardiac rupture after myocardial infarction in MI mice. High-dose metformin caused cardiac rupture in MI mice. Moreover, high-dose metformin (Met 2.0 nM) reduces the proportion of NT-PGC-1α/PGC-1α in primary cardiomyocytes of SD mice (SD-NRVCs [Neonatal rat ventricular cardiomyocytes]), and its effect was inhibited by Compound C (AMPK inhibitor). Further, after 3 days of treatment with high-dose metformin in MI mice, myocardial fibrin synthesis decreased and fibrosis was significantly inhibited. Meanwhile, cardiomyocyte apoptosis increased significantly. With the increase in metformin concentration, the expression level of myocardial LC3b gradually increased in MI mice, suggesting that metformin enhances the autophagy of cardiomyocytes. Conclusions These results suggest that metformin increases cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Jinghai Hua
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Zhanghua Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Dongqi An
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Wenyan Lai
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland).,Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
144
|
Das S, Behera SK, Srinivasan A, Xavier AS, Selvarajan S, Kamalanathan S, Sahoo JP, Nair NS. Effect of metformin on exercise capacity: A meta-analysis. Diabetes Res Clin Pract 2018; 144:270-278. [PMID: 30217594 DOI: 10.1016/j.diabres.2018.08.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022]
Abstract
AIMS To evaluate the effect of metformin on various parameters of exercise capacity [oxygen consumption (VO2), peak oxygen consumption (VO2peak), heart rate (HR), exercise test duration, respiratory exchange ratio (RER), rating of perceived exertion (RPE), lactate and ventilatory anaerobic threshold (VAT)]. METHODS Studies reporting change in VO2 or VO2peak after metformin administration were included. Subgroup analyses were performed as applicable. Mean difference with 95% CIs were pooled using random-effects model [RevMan (v5.3)]. RESULTS There were no changes in VO2 and VO2peak in the overall population [VO2: n = 388, mean difference: -0.12 ml/kg/min, 95% CI: -0.74, 0.51, p = 0.71 (i2 = 0%, p = 0.99); VO2peak: n = 345, mean difference: 0.41 ml/kg/min, 95% CI: -0.51, 1.33, p = 0.38 (i2 = 0%, p = 0.89)], healthy volunteers and patients (type 2 diabetes mellitus, insulin resistance, impaired glucose tolerance/impaired fasting glucose and metabolic syndrome). For patients with insulin resistance, there was a decrease in VO2peak, but not VO2. In the overall population, there was a significant decrease in HR and RER, a significant increase in RPE, and no changes in exercise test duration and VAT. In addition, there was an increased VAT in the healthy volunteers. CONCLUSIONS In the overall population, metformin did not affect VO2, VO2peak, exercise test duration and VAT, although it significantly decreased HR, RER and increased RPE.
Collapse
Affiliation(s)
- Saibal Das
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sapan Kumar Behera
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Akila Srinivasan
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Alphienes Stanley Xavier
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Sandhiya Selvarajan
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India.
| | - Sadishkumar Kamalanathan
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Jaya Prakash Sahoo
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - N Sreekumaran Nair
- Department of Biostatistics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
145
|
Piskovatska V, Stefanyshyn N, Storey KB, Vaiserman AM, Lushchak O. Metformin as a geroprotector: experimental and clinical evidence. Biogerontology 2018; 20:33-48. [PMID: 30255224 DOI: 10.1007/s10522-018-9773-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Apart from being a safe, effective and globally affordable glucose-lowering agent for the treatment of diabetes, metformin has earned much credit in recent years as a potential anti-aging formula. It has been shown to significantly increase lifespan and delay the onset of age-associated decline in several experimental models. The current review summarizes advances in clinical research on the potential role of metformin in the field of geroprotection, highlighting findings from pre-clinical studies on known and putative mechanisms behind its beneficial properties. A growing body of evidence from clinical trials demonstrates that metformin can effectively reduce the risk of many age-related diseases and conditions, including cardiometabolic disorders, neurodegeneration, cancer, chronic inflammation, and frailty. Metformin also holds promise as a drug that could be repurposed for chemoprevention or adjuvant therapy for certain cancer types. Moreover, due to the ability of metformin to induce autophagy by activation of AMPK, it is regarded as a potential hormesis-inducing agent with healthspan-promoting and pro-longevity properties. Long-term intake of metformin is associated with low risk of adverse events; however, well-designed clinical trials are still warranted to enable potential use of this therapeutic agent as a geroprotector.
Collapse
Affiliation(s)
- Veronika Piskovatska
- Clinic for Heart Surgery, University Clinic of the Martin Luther University, Halle, Germany
| | - Nadiya Stefanyshyn
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | | | | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine.
| |
Collapse
|
146
|
Genkel VV, Nikushkina KV, Nikonova TI, Shaposhnik II. Peripheral arterial disease and indicators of low-grade inflammation in patients with coronary artery disease and type 2 diabetes mellitus. DIABETES MELLITUS 2018. [DOI: 10.14341/dm9392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND: The study of low-grade inflammation in patients with type 2 diabetes mellitus (T2DM) and atherosclerotic cardiovascular diseases is a pressing problem. A deeper understanding of the cascade of inflammatory reactions, possibly mediating the severe atherosclerotic lesions of various vascular pools in patients with diabetes, has the potential to introduce more sophisticated diagnostic and therapeutic approaches into practice.
AIM: To study the interrelation of low-grade inflammation and atherosclerosis of peripheral arteries in patients with coronary artery disease (CAD) and T2DM.
MATERIALS AND METHODS: The study included 137 patients (77 men and 60 women) with CAD. The average age of patients was 62.0 (57.066.0) years. The first group included 67 patients with CAD and T2DM, and the second group included 70 patients with CAD. Low-grade inflammation was assessed by the levels of high-sensitivity C-reactive protein, interleukin (IL)-1, IL-6, IL-8, IL-10 and TNF-. All patients underwent duplex scanning of carotid arteries and lower extremity arteries (LEAs).
RESULTS: Patients with CAD and T2DM showed significantly greater values of stenosis of carotid arteries and LEAs. Direct correlation was revealed between markers of inflammation and the degree of stenosis of the femoral and tibial arteries, as well as the intima-media thickness of the carotid and femoral arteries. In the group of patients with T2DM, the value of IL-1 was 2.04 (0.982.52) pg/mL, which was significantly less than 2.43 (1.843.19) pg/mL for patients in the second group (p = 0.010). The values of IL-6 were also significantly lower in the first group of patients, at 1.84 (0.734.41) pg/mL vs. 3.73 (2.2710.2) pg/mL in the first and second groups, respectively (p = 0.008). The dose of metformin was inversely correlated with the level of IL-6 (r = 0.314, p = 0.003).
CONCLUSIONS: Patients with CAD and T2DM compared with patients without diabetes had significantly greater values of stenosis of peripheral arteries. The levels of IL-1 and IL-6 in the group of patients with CAD and T2DM were significantly lower in comparison with patients without diabetes. The dose of metformin was inversely correlated with the level of IL-6.
Collapse
|
147
|
Tseng CH. Metformin and Risk of Hypertension in Taiwanese Patients With Type 2 Diabetes Mellitus. J Am Heart Assoc 2018; 7:e008860. [PMID: 29954747 PMCID: PMC6064894 DOI: 10.1161/jaha.118.008860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/09/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Whether metformin use may reduce hypertension risk has not been studied. This study investigated such possibility in patients with type 2 diabetes mellitus. METHODS AND RESULTS Newly diagnosed patients with type 2 diabetes mellitus during 1999-2005 were enrolled from the reimbursement database of the Taiwan's National Health Insurance and followed to December 31, 2011. Hypertension was defined either by a diagnosis or by a diagnosis plus the use of angiotensin converting enzyme inhibitors/angiotensin receptor blockers and/or calcium channel blockers. Analyses were conducted in a propensity score matched-pair cohort of 4810 ever users and 4810 never users. Cox proportional hazards regression model was used to estimate the hazard ratios. Results showed that when hypertension was defined by a diagnosis, 2261 never users and 1908 ever users developed hypertension. The overall hazard ratio was 0.724 (0.681-0.769) and the hazard ratios for the first (<2.0 months), second (2.0-13.0 months) and third (>13.0 months) tertiles of cumulative duration were 0.820 (0.745-0.903), 0.692 (0.634-0.756), and 0.687 (0.630-0.749), respectively. When cumulative duration of metformin therapy was treated as a continuous variable, the hazard ratio was 0.991 (0.989-0.994) for every 1-month increment of metformin use. When hypertension was defined by a diagnosis plus the use of antihypertensive drugs, the overall hazard ratio was 0.831 (0.771-0.895), the hazard ratios for the respective tertiles were 0.868 (0.769-0.980), 0.852 (0.767-0.946), and 0.787 (0.709-0.874), and the hazard ratio was 0.994 (0.991-0.997) for every 1-month increment of metformin use. CONCLUSIONS A reduced risk of hypertension is observed in metformin users in a dose-response pattern.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
148
|
Bala C, Rusu A, Ciobanu DM, Craciun AE, Roman G. The association study of high-sensitivity C-reactive protein, pentraxin 3, nitrotyrosine, and insulin dose in patients with insulin-treated type 2 diabetes mellitus. Ther Clin Risk Manag 2018; 14:955-963. [PMID: 29881277 PMCID: PMC5978465 DOI: 10.2147/tcrm.s162086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE The objective of this study was to examine the association between insulin dose and high-sensitivity C-reactive protein (hsCRP), nitrotyrosine, and pentraxin 3 in patients with insulin-treated type 2 diabetes. PATIENTS AND METHODS Eighty patients with type 2 diabetes treated with insulin for >6 months and with stable insulin doses (±10%) within 3 months before inclusion were enrolled in this study. Medical history, including use of insulin and insulin doses, concomitant diseases and medication, and anthropometric and routine biochemical parameters were collected for each patient. hsCRP, nitrotyrosine, and pentraxin 3 were measured in fasting conditions. Comparison analysis was performed according to the distribution in tertiles of insulin dose/kg of body weight, and linear regression adjusted for confounding factors was used to examine the associations between markers of inflammation, oxidative stress, and insulin dose. RESULTS In the comparison analysis, no statistically significant difference was found between hsCRP, nitrotyrosine, and pentraxin 3 levels across tertiles of insulin dose expressed as IU/kg of body weight (p for trend >0.05 for all comparisons) except a significantly higher hsCRP level in tertile 3 compared to tertile 1 (3.9±3.6 vs 6.1±3.8 mg/dL, p=0.035). In regression analysis, after adjustment for age, gender, smoking, body mass index, glycated hemoglobin, C-peptide, metformin, antiplatelet, and statin use, only hsCRP levels were statistically significant associated with insulin dose/kg of body weight (β=0.237, p=0.043). CONCLUSION In this sample of patients with type 2 diabetes treated with insulin for >6 months, hsCRP was positively associated with insulin doses. No such association was found for pentraxin 3, a more specific marker of vascular inflammation, and for nitrotyrosine as a marker of oxidative stress.
Collapse
Affiliation(s)
- Cornelia Bala
- Department of Diabetes and Nutrition, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adriana Rusu
- Department of Diabetes and Nutrition, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dana Mihaela Ciobanu
- Department of Diabetes and Nutrition, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Elena Craciun
- Department of Diabetes and Nutrition, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriela Roman
- Department of Diabetes and Nutrition, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
149
|
Mantuano P, Sanarica F, Conte E, Morgese MG, Capogrosso RF, Cozzoli A, Fonzino A, Quaranta A, Rolland JF, De Bellis M, Camerino GM, Trabace L, De Luca A. Effect of a long-term treatment with metformin in dystrophic mdx mice: A reconsideration of its potential clinical interest in Duchenne muscular dystrophy. Biochem Pharmacol 2018; 154:89-103. [PMID: 29684379 DOI: 10.1016/j.bcp.2018.04.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 12/18/2022]
Abstract
The pharmacological stimulation of AMP-activated protein kinase (AMPK) via metabolic enhancers has been proposed as potential therapeutic strategy for Duchenne muscular dystrophy (DMD). Metformin, a widely-prescribed anti-hyperglycemic drug which activates AMPK via mitochondrial respiratory chain, has been recently tested in DMD patients in synergy with nitric oxide (NO)-precursors, with encouraging results. However, preclinical data supporting the use of metformin in DMD are still poor, and its actions on skeletal muscle appear controversial. Therefore, we investigated the effects of a long-term treatment with metformin (200 mg/kg/day in drinking water, for 20 weeks) in the exercised mdx mouse model, characterized by a severe mechanical-metabolic maladaptation. Metformin significantly ameliorated histopathology in mdx gastrocnemius muscle, in parallel reducing TGF-β1 with a recovery score (r.s) of 106%; this was accompanied by a decreased plasma matrix-metalloproteinase-9 (r.s. 43%). In addition, metformin significantly increased mdx diaphragm twitch and tetanic tension ex vivo (r.s. 44% and 36%, respectively), in spite of minor effects on in vivo weakness. However, no clear protective actions on dystrophic muscle metabolism were observed, as shown by the poor metformin effect on AMPK activation measured by western blot, on the expression of mechanical-metabolic response genes analyzed by qPCR, and by the lack of fast-to-slow fiber-type-shift assessed by SDH staining in tibialis anterior muscle. Similar results were obtained in the milder phenotype of sedentary mdx mice. The lack of metabolic effects could be, at least partly, due to metformin inability to increase low mdx muscle levels of l-arginine, l-citrulline and taurine, found by HPLC. Our findings encourage to explore alternative, metabolism-independent mechanisms of action to differently repurpose metformin in DMD, supporting its therapeutic combination with NO-sources.
Collapse
Affiliation(s)
- Paola Mantuano
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Francesca Sanarica
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Elena Conte
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Maria Grazia Morgese
- Department of Experimental and Clinical Medicine, Faculty of Medicine, University of Foggia, Foggia, Italy
| | | | - Anna Cozzoli
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Adriano Fonzino
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Angelo Quaranta
- Department of Veterinary Medicine, University of Bari "Aldo Moro", Valenzano, Bari, Italy
| | | | - Michela De Bellis
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Giulia Maria Camerino
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Luigia Trabace
- Department of Experimental and Clinical Medicine, Faculty of Medicine, University of Foggia, Foggia, Italy
| | - Annamaria De Luca
- Section of Pharmacology, Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
150
|
Tain YL, Wu KLH, Lee WC, Leu S, Chan JYH. Prenatal Metformin Therapy Attenuates Hypertension of Developmental Origin in Male Adult Offspring Exposed to Maternal High-Fructose and Post-Weaning High-Fat Diets. Int J Mol Sci 2018; 19:ijms19041066. [PMID: 29614026 PMCID: PMC5979307 DOI: 10.3390/ijms19041066] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 03/19/2018] [Accepted: 04/01/2018] [Indexed: 01/04/2023] Open
Abstract
Widespread consumption of a Western diet, comprised of highly refined carbohydrates and fat, may play a role in the epidemic of hypertension. Hypertension can take origin from early life. Metformin is the preferred treatment for type 2 diabetes. We examined whether prenatal metformin therapy can prevent maternal high-fructose plus post-weaning high-fat diets-induced hypertension of developmental origins via regulation of nutrient sensing signals, uric acid, oxidative stress, and the nitric oxide (NO) pathway. Gestating Sprague–Dawley rats received regular chow (ND) or chow supplemented with 60% fructose diet (HFR) throughout pregnancy and lactation. Male offspring were onto either the ND or high-fat diet (HFA) from weaning to 12 weeks of age. A total of 40 male offspring were assigned to five groups (n = 8/group): ND/ND, HFR/ND, ND/HFA, HFR/HFA, and HFR/HFA+metformin. Metformin (500 mg/kg/day) was administered via gastric gavage for three weeks during the pregnancy period. Combined maternal HFR plus post-weaning HFA induced hypertension in male adult offspring, which prenatal metformin therapy prevented. The protective effects of prenatal metformin therapy on HFR/HFA-induced hypertension, including downregulation of the renin-angiotensin system, decrease in uric acid level, and reduction of oxidative stress. Our results highlighted that the programming effects of metformin administered prenatally might be different from those reported in adults, and that deserves further elucidation.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Kay L H Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Wei-Chia Lee
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Steve Leu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| |
Collapse
|