401
|
Elbere I, Silamikelis I, Ustinova M, Kalnina I, Zaharenko L, Peculis R, Konrade I, Ciuculete DM, Zhukovsky C, Gudra D, Radovica-Spalvina I, Fridmanis D, Pirags V, Schiöth HB, Klovins J. Significantly altered peripheral blood cell DNA methylation profile as a result of immediate effect of metformin use in healthy individuals. Clin Epigenetics 2018; 10:156. [PMID: 30545422 PMCID: PMC6293577 DOI: 10.1186/s13148-018-0593-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Metformin is a widely prescribed antihyperglycemic agent that has been also associated with multiple therapeutic effects in various diseases, including several types of malignancies. There is growing evidence regarding the contribution of the epigenetic mechanisms in reaching metformin's therapeutic goals; however, the effect of metformin on human cells in vivo is not comprehensively studied. The aim of our study was to examine metformin-induced alterations of DNA methylation profiles in white blood cells of healthy volunteers, employing a longitudinal study design. RESULTS Twelve healthy metformin-naïve individuals where enrolled in the study. Genome-wide DNA methylation pattern was estimated at baseline, 10 h and 7 days after the start of metformin administration. The whole-genome DNA methylation analysis in total revealed 125 differentially methylated CpGs, of which 11 CpGs and their associated genes with the most consistent changes in the DNA methylation profile were selected: POFUT2, CAMKK1, EML3, KIAA1614, UPF1, MUC4, LOC727982, SIX3, ADAM8, SNORD12B, VPS8, and several differentially methylated regions as novel potential epigenetic targets of metformin. The main functions of the majority of top-ranked differentially methylated loci and their representative cell signaling pathways were linked to the well-known metformin therapy targets: regulatory processes of energy homeostasis, inflammatory responses, tumorigenesis, and neurodegenerative diseases. CONCLUSIONS Here we demonstrate for the first time the immediate effect of short-term metformin administration at therapeutic doses on epigenetic regulation in human white blood cells. These findings suggest the DNA methylation process as one of the mechanisms involved in the action of metformin, thereby revealing novel targets and directions of the molecular mechanisms underlying the various beneficial effects of metformin. TRIAL REGISTRATION EU Clinical Trials Register, 2016-001092-74. Registered 23 March 2017, https://www.clinicaltrialsregister.eu/ctr-search/trial/2016-001092-74/LV .
Collapse
Affiliation(s)
- Ilze Elbere
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ivars Silamikelis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Monta Ustinova
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ineta Kalnina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Linda Zaharenko
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Raitis Peculis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Konrade
- Riga East Clinical University Hospital, 2 Hipokrata Street, Riga, LV-1038, Latvia
| | - Diana Maria Ciuculete
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Christina Zhukovsky
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Dita Gudra
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Radovica-Spalvina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Davids Fridmanis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia.
| |
Collapse
|
402
|
Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL, Divers J, Espeland MA, Marcovina S, Pollak MN, Kritchevsky SB, Barzilai N, Kuchel GA. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. GeroScience 2018; 40:419-436. [PMID: 30151729 PMCID: PMC6294728 DOI: 10.1007/s11357-018-0042-y] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Recent advances indicate that biological aging is a potentially modifiable driver of late-life function and chronic disease and have led to the development of geroscience-guided therapeutic trials such as TAME (Targeting Aging with MEtformin). TAME is a proposed randomized clinical trial using metformin to affect molecular aging pathways to slow the incidence of age-related multi-morbidity and functional decline. In trials focusing on clinical end-points (e.g., disease diagnosis or death), biomarkers help show that the intervention is affecting the underlying aging biology before sufficient clinical events have accumulated to test the study hypothesis. Since there is no standard set of biomarkers of aging for clinical trials, an expert panel was convened and comprehensive literature reviews conducted to identify 258 initial candidate biomarkers of aging and age-related disease. Next selection criteria were derived and applied to refine this set emphasizing: (1) measurement reliability and feasibility; (2) relevance to aging; (3) robust and consistent ability to predict all-cause mortality, clinical and functional outcomes; and (4) responsiveness to intervention. Application of these selection criteria to the current literature resulted in a short list of blood-based biomarkers proposed for TAME: IL-6, TNFα-receptor I or II, CRP, GDF15, insulin, IGF1, cystatin C, NT-proBNP, and hemoglobin A1c. The present report provides a conceptual framework for the selection of blood-based biomarkers for use in geroscience-guided clinical trials. This work also revealed the scarcity of well-vetted biomarkers for human studies that reflect underlying biologic aging hallmarks, and the need to leverage proposed trials for future biomarker discovery and validation.
Collapse
Affiliation(s)
- Jamie N Justice
- Internal Medicine Section on Gerontology and Geriatrics, and the Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Vanita R Aroda
- Department of Medicine, Division of Diabetes, Endocrinology, and Hypertension Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Judy L Bahnson
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jasmin Divers
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mark A Espeland
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Santica Marcovina
- Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Michael N Pollak
- Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec, H3T1E2, Canada
| | - Stephen B Kritchevsky
- Internal Medicine Section on Gerontology and Geriatrics, and the Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Nir Barzilai
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| |
Collapse
|
403
|
Logie L, Lees Z, Allwood JW, McDougall G, Beall C, Rena G. Regulation of hepatic glucose production and AMPK by AICAR but not by metformin depends on drug uptake through the equilibrative nucleoside transporter 1 (ENT1). Diabetes Obes Metab 2018; 20:2748-2758. [PMID: 29962100 PMCID: PMC6282725 DOI: 10.1111/dom.13455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 01/15/2023]
Abstract
AIM Recently we have observed differences in the ability of metformin and AICAR to repress glucose production from hepatocytes using 8CPT-cAMP. Previous results indicate that, in addition to activating protein kinase A, 8CPT-modified cAMP analogues suppress the nitrobenzylthioinosine (NBMPR)-sensitive equilibrative nucleoside transporter ENT1. We aimed to exploit 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR, which is highly selective for a high-affinity binding-site on ENT1, to investigate the role of ENT1 in the liver-specific glucose-lowering properties of AICAR and metformin. METHODS Primary mouse hepatocytes were incubated with AICAR and metformin in combination with cAMP analogues, glucagon, forskolin and NBMPR. Hepatocyte glucose production (HGP) and AMPK signalling were measured, and a uridine uptake assay with supporting LC-MS was used to investigate nucleoside depletion from medium by cells. RESULTS AICAR and metformin increased AMPK pathway phosphorylation and decreased HGP induced by dibutyryl cAMP and glucagon. HGP was also induced by 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR; however, in each case this was resistant to suppression by AICAR but not by metformin. Cross-validation of tracer and mass spectrometry studies indicates that 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR inhibited the effects of AICAR, at least in part, by impeding its uptake into hepatocytes. CONCLUSIONS We report for the first time that suppression of ENT1 induces HGP. ENT1 inhibition also impedes uptake and the effects of AICAR, but not metformin, on HGP. Further investigation of nucleoside transport may illuminate a better understanding of how metformin and AICAR each regulate HGP.
Collapse
Affiliation(s)
- Lisa Logie
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
| | - Zoe Lees
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - J. William Allwood
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - Gordon McDougall
- Environmental and Biochemical SciencesThe James Hutton InstituteDundeeUK
| | - Craig Beall
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| | - Graham Rena
- Division of Cellular MedicineNinewells Hospital and Medical School, University of DundeeDundeeUK
| |
Collapse
|
404
|
Prattichizzo F, Giuliani A, Mensà E, Sabbatinelli J, De Nigris V, Rippo MR, La Sala L, Procopio AD, Olivieri F, Ceriello A. Pleiotropic effects of metformin: Shaping the microbiome to manage type 2 diabetes and postpone ageing. Ageing Res Rev 2018; 48:87-98. [PMID: 30336272 DOI: 10.1016/j.arr.2018.10.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/13/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023]
Abstract
Metformin is the first-choice therapy to lower glycaemia and manage type 2 diabetes. Continuously emerging epidemiological data and experimental models are showing additional protective effects of metformin against a number of age-related diseases (ARDs), e.g., cardiovascular diseases and cancer. This evidence has prompted the design of a specific trial, i.e., the Targeting Aging with Metformin (TAME) trial, to test metformin as an anti-ageing molecule. However, a unifying or prevailing mechanism of action of metformin is still debated. Here, we summarize the epidemiological data linking metformin to ARD prevention. Then, we dissect the deeply studied mechanisms of action explaining its antihyperglycemic effect and the putative mechanisms supporting its anti-ageing properties, focusing on studies using clinically pertinent doses. We hypothesize that the molecular observations obtained in different models with metformin could be indirectly mediated by its effect on gut flora. Novel evidence suggests that metformin reshapes the human microbiota, promoting the growth of beneficial bacterial species and counteracting the expansion of detrimental bacterial species. In turn, this action would influence the balance between pro- and anti-inflammatory circulating factors, thereby promoting glycaemic control and healthy ageing. This framework may reconcile diverse observations, providing information for designing further studies to elucidate the complex interplay between metformin and the metabiome harboured in mammalian body compartments, thereby paving the way for innovative, bacterial-based therapeutics to manage type 2 diabetes and foster a longer healthspan.
Collapse
Affiliation(s)
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Emanuela Mensà
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Valeria De Nigris
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, Italian National Research Centre on Aging, IRCCS INRCA, Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, Italian National Research Centre on Aging, IRCCS INRCA, Ancona, Italy
| | - Antonio Ceriello
- IRCCS MultiMedica, Milan, Italy; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| |
Collapse
|
405
|
Yaribeygi H, Atkin SL, Pirro M, Sahebkar A. A review of the anti-inflammatory properties of antidiabetic agents providing protective effects against vascular complications in diabetes. J Cell Physiol 2018; 234:8286-8294. [PMID: 30417367 DOI: 10.1002/jcp.27699] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
The global prevalence of Type 2 diabetes mellitus and its associated complications are growing rapidly. Although the role of hyperglycemia is well recognized in the pathophysiology of diabetic complications, its exact underlying mechanisms are not fully understood. In this regard, accumulating evidence suggests that the role of inflammation appears pivotal, with studies showing that most diabetic complications are associated with an inflammatory response. Several classes of antidiabetic agents have been introduced for controlling glycemia, with evidence that these pharmacological agents may have modulatory effects on inflammation beyond their glucose-lowering activity. Here we review the latest evidence on the anti-inflammatory effects of commonly used antidiabetic medications and discuss the relevance of these effects on preventing diabetic complications.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Matteo Pirro
- Department of Medicine, Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, University of Perugia, Perugia, Italy
| | - 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
|
406
|
Satoh K, Shimokawa H. Sirtuin 5 promotes ischemia/reperfusion-induced blood-brain barrier damage after stroke. Int J Cardiol 2018; 284:77-78. [PMID: 30448019 DOI: 10.1016/j.ijcard.2018.11.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
407
|
Abdellatif M, Sedej S, Madeo F, Kroemer G. Cardioprotective effects of autophagy induction in sepsis. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:S61. [PMID: 30613636 PMCID: PMC6291539 DOI: 10.21037/atm.2018.10.23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/11/2018] [Indexed: 01/12/2023]
Affiliation(s)
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Frank Madeo
- BioTechMed Graz, Graz, Austria
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- INSERM, U1138, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
408
|
Wang SQ, Cui SX, Qu XJ. Metformin inhibited colitis and colitis-associated cancer (CAC) through protecting mitochondrial structures of colorectal epithelial cells in mice. Cancer Biol Ther 2018; 20:338-348. [PMID: 30359174 DOI: 10.1080/15384047.2018.1529108] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although a mountain of papers have showed that metformin plays a role in inhibiting cancers, but the mechanism underpinning this has not yet fully elucidated. Herein, we used AOM/DSS model, the clinicopathological features are similar to those found in humans, to investigate the effects of metformin as well as combination with 5-FU in the prevention of colitis and colitis associated cancer (CAC). Oral metformin significantly inhibited DSS-induced ulcerative colitis and AOM/DSS-induced CAC. Metformin also ameliorated 5-FU-induced colorectal gastrointestinal symptoms in mice. Metformin combination with 5-FU strongly inhibited colorectal cancer. Metformin reduced levels of the NFκB signaling components p-IKKα/β, p-NFκB, p-IκBα in colorectal mucosal cells. Transmission electron microscopy analysis suggested that the inhibition of metformin on colitis and CAC might associate with its biological activity of protecting mitochondrial structures of colorectal epithelial cells. Further analysis by Mito Tracker Red staining assay indicated that metformin prevented H2O2-induced mitochondrial fission correlated with a decrease of mitochondrial perimeter. In addition, metformin increased the level of NDUFA9, a Q-module subunit required for complex I assembly, in colorectal epithelial cells. These observations of metformin in the inhibition of colitis and CAC might associate with its activity of activating the LKB1/AMPK pathway in colorectal epithelial cells. In conclusion, metformin inhibited colitis and CAC through protecting the mitochondrial structures of colorectal epithelial cells.
Collapse
Affiliation(s)
- Shu-Qing Wang
- a Department of Pharmacology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Shu-Xiang Cui
- b Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , China
| | - Xian-Jun Qu
- a Department of Pharmacology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| |
Collapse
|
409
|
Wang HC, Huang SK. Metformin inhibits IgE- and aryl hydrocarbon receptor-mediated mast cell activation in vitro and in vivo. Eur J Immunol 2018; 48:1989-1996. [PMID: 30242842 DOI: 10.1002/eji.201847706] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/31/2018] [Accepted: 09/20/2018] [Indexed: 01/15/2023]
Abstract
Metformin, an anti-diabetic drug, possesses anti-inflammatory property beyond its glucose-lowering activity, but its regulatory effect on mast cells and allergic responses remains unknown, wherein the aryl hydrocarbon receptor (AhR)-ligand axis is critical in controlling mast cell activation. Herein, we provide evidence supporting the role of metformin in modulating mast cell activation by FcεR1-, AhR-mediated signaling or their combination. Metformin at relatively low doses was shown to suppress FcεR1-mediated degranulation, IL-13, TNF-α and sphingosine-1-phosphate (S1P) secretion in murine bone marrow-derived mast cells (BMMCs). In contrast, metformin at the same doses potently inhibited all parameters in mast cells stimulated with an AhR ligand, 5,11-dihydroindolo[3,2-b]carbazole-6-carbaldehyde (FICZ). Further, metformin was shown to inhibit FcεR1- and AhR-mediated passive cutaneous anaphylaxis (PCA) in vivo, reversible by a S1P receptor 2 antagonist, JTE-013. Using AhR reporter cells, Huh7-DRE-Luc cells, a human mast cell line, HMC-1, and BMMCs, metformin's inhibitory effect was mediated through the suppression of FICZ-induced AhR activity, calcium mobilization and ROS generation. Notably, FICZ-mediated oxidation of S1P lyase (S1PL) and its reduced activity were reversed by metformin, resulting in decreased levels of S1P. Collectively, these results suggested the potential utility of metformin in treating allergic diseases, particularly in cases with comorbid type II diabetes mellitus.
Collapse
Affiliation(s)
- Hsueh-Chun Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan.,Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Lou-Hu Hospital, Shen-Zhen University, Shen-Zhen, China
| |
Collapse
|
410
|
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: 11.0] [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
|
411
|
Zhao Q, Zhou X, Curbo S, Karlsson A. Metformin downregulates the mitochondrial carrier SLC25A10 in a glucose dependent manner. Biochem Pharmacol 2018; 156:444-450. [PMID: 30222970 DOI: 10.1016/j.bcp.2018.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Metformin, a commonly used agent in the treatment of type 2 diabetes, is also associated with reduced risk of cancer development and improvement in cancer survival. Although much is known about metformin, the mechanisms behind its anti-cancer properties are not fully understood. In this study we addressed the role of a mitochondrial transporter commonly upregulated in cancer cells, SLC25A10, for cell survival and metabolism in the presence of metformin. SLC25A10 is a carrier in the mitochondrial inner membrane that transports malate and succinate out of the mitochondria, in exchange of phosphate and sulfate. We show that metformin treatment results in decreased gene expression of the SLC25A10 carrier both in lung cancer A549 mock cells and A549 SLC25A10 knockdown (siSLC25A10) cells. The decrease was even more pronounced when cells were grown at low glucose concentrations. The expression levels of key enzymes in glucose metabolism showed slightly altered mean values for all genes tested in both control cells and siSLC25A10 cells upon metformin treatment. The gene expression of the metabolic regulator glutamic-oxaloacetic transaminase 1 decreased in wild type cells upon metformin treatment whereas there was a trend of increased expression in the siSLC25A10 cells upon metformin treatment. In addition, the gene expression of the cyclin-dependent kinase inhibitor 1A was markedly increased in the siSLC25A10 compared to control A549 cells, and with even larger increases in the presence of metformin and at low glucose concentration. Our data show that in siSLC25A10 cell lines, metformin significantly alters the SLC25A10 carrier at both mRNA and protein levels and can thereby affect the supply of nutrients and the metabolic state of cancer cells.
Collapse
Affiliation(s)
- Qian Zhao
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Xiaoshan Zhou
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Sophie Curbo
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Anna Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, 141 86 Stockholm, Sweden
| |
Collapse
|
412
|
Saxagliptin regulates M1/M2 macrophage polarization via CaMKKβ/AMPK pathway to attenuate NAFLD. Biochem Biophys Res Commun 2018; 503:1618-1624. [DOI: 10.1016/j.bbrc.2018.07.090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/19/2018] [Indexed: 01/13/2023]
|
413
|
Jia Y, Han Y, Wang X, Han F. Role of apoptosis in the Post-traumatic stress disorder model-single prolonged stressed rats. Psychoneuroendocrinology 2018; 95:97-105. [PMID: 29843020 DOI: 10.1016/j.psyneuen.2018.05.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a stress-related mental disorder which occurs following exposure to traumatic events. A number of brain neuroimaging studies have revealed that PTSD patients have reduced volume and abnormal functions in the hippocampus and the amygdala. However, the pathogenesis of abnormalities in certain brain regions, as induced by PTSD, remains unclear. Recent studies, using the single prolonged stress (SPS) model, an animal model of PTSD, have found that abnormal apoptosis in certain brain regions, including the hippocampus, the amygdala, and the medial prefrontal cortex (mPFC); these areas are closely associated with emotion and cognition. In this review, we summarize the mechanism of apoptosis in SPS rats, including the endoplasmic reticulum (ER) and the mitochondria pathways. For the ER pathway, three individual pathways: PERK, IRE1, and ATF6 showed different roles on apoptosis and neuroprotection. Three key factors are thought to be involved in the mitochondrial pathway and PTSD-induced apoptosis: corticosteroid receptors, apoptosis-related factors, and anti-apoptosis factors. We have investigated the role of these factors and have attempted to identify which factors of the pathways are more focused towards neuronal protection, and which are more direct towards apoptosis. We also discussed the role of autophagy and the specific differences between autophagy and apoptosis in SPS rats. Finally, we discussed emerging researches related to anti-apoptosis treatment, including PERK inhibitors, IRE1 inhibitors, and metformin; collectively, these were exciting, but limited, This review provides a summary of the current understanding of apoptosis in SPS rats and the potential anti-apoptosis treatment strategies for PTSD.
Collapse
Affiliation(s)
- Yunbo Jia
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Yunhe Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Xinyue Wang
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Fang Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
414
|
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: 0.5] [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
|
415
|
Resolution of chronic inflammatory disease: universal and tissue-specific concepts. Nat Commun 2018; 9:3261. [PMID: 30111884 PMCID: PMC6093916 DOI: 10.1038/s41467-018-05800-6] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammation and its resolution is under-studied in medicine despite being essential for understanding the development of chronic inflammatory disease. In this review article, we discuss the resolution of inflammation in both a biological and translational context. We introduce the concept of impaired resolution leading to diseases like rheumatoid arthritis, Crohn's disease, and asthma, as well as the cellular and molecular components that contribute to resolution of joint, gut, and lung inflammation, respectively. Finally, we discuss potential intervention strategies for fostering the resolution process, and their implications for the therapy of inflammatory diseases.
Collapse
|
416
|
Maniar K, Singh V, Moideen A, Bhattacharyya R, Chakrabarti A, Banerjee D. Inhalational supplementation of metformin butyrate: A strategy for prevention and cure of various pulmonary disorders. Biomed Pharmacother 2018; 107:495-506. [PMID: 30114633 DOI: 10.1016/j.biopha.2018.08.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/22/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022] Open
Abstract
The management of chronic lung diseases such as cancer, asthma, COPD and pulmonary hypertension remains unsatisfactory till date, and several strategies are being tried to control the same. Metformin, a popular anti-diabetic drug has shown promising effects in pre-clinical studies and has been subject to several trials in patients with debilitating pulmonary diseases. However, the clinical evidence for the use of metformin in these conditions is disappointing. Recent observations suggest that metformin use in diabetic patients is associated with an increase in butyrate-producing bacteria in the gut microbiome. Butyrate, similar to metformin, shows beneficial effects in pathological conditions found in pulmonary diseases. Further, the pharmacokinetic data of metformin suggests that metformin is predominantly concentrated in the gut, even after absorption. Butyrate, on the other hand, has a short half-life and thus oral supplementation of butyrate and metformin is unlikely to result in high concentrations of these drugs in the lung. In this paper, we review the pre-clinical studies of metformin and butyrate pertaining to pathologies commonly encountered in chronic lung diseases and underscore the need to administer these drugs directly to the lung via the inhalational route.
Collapse
Affiliation(s)
- Kunal Maniar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, India
| | - Vandana Singh
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, India
| | - Amal Moideen
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, India
| | - Rajasri Bhattacharyya
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, India
| | - Amitava Chakrabarti
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, India
| | - Dibyajyoti Banerjee
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, India.
| |
Collapse
|
417
|
Metformin treatment prevents gallstone formation but mimics porcelain gallbladder in C57Bl/6 mice. Eur J Pharmacol 2018; 833:165-172. [DOI: 10.1016/j.ejphar.2018.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 11/15/2022]
|
418
|
Nikolakopoulou P, Chatzigeorgiou A, Kourtzelis I, Toutouna L, Masjkur J, Arps-Forker C, Poser SW, Rozman J, Rathkolb B, Aguilar-Pimentel JA, Wolf E, Klingenspor M, Ollert M, Schmidt-Weber C, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Tsata V, Monasor LS, Troullinaki M, Witt A, Anastasiou V, Chrousos G, Yi CX, García-Cáceres C, Tschöp MH, Bornstein SR, Androutsellis-Theotokis A. Streptozotocin-induced β-cell damage, high fat diet, and metformin administration regulate Hes3 expression in the adult mouse brain. Sci Rep 2018; 8:11335. [PMID: 30054579 PMCID: PMC6063949 DOI: 10.1038/s41598-018-29434-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 07/09/2018] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus is a group of disorders characterized by prolonged high levels of circulating blood glucose. Type 1 diabetes is caused by decreased insulin production in the pancreas whereas type 2 diabetes may develop due to obesity and lack of exercise; it begins with insulin resistance whereby cells fail to respond properly to insulin and it may also progress to decreased insulin levels. The brain is an important target for insulin, and there is great interest in understanding how diabetes affects the brain. In addition to the direct effects of insulin on the brain, diabetes may also impact the brain through modulation of the inflammatory system. Here we investigate how perturbation of circulating insulin levels affects the expression of Hes3, a transcription factor expressed in neural stem and progenitor cells that is involved in tissue regeneration. Our data show that streptozotocin-induced β-cell damage, high fat diet, as well as metformin, a common type 2 diabetes medication, regulate Hes3 levels in the brain. This work suggests that Hes3 is a valuable biomarker helping to monitor the state of endogenous neural stem and progenitor cells in the context of diabetes mellitus.
Collapse
Affiliation(s)
| | - Antonios Chatzigeorgiou
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Ioannis Kourtzelis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Louiza Toutouna
- Department of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jimmy Masjkur
- Department of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Carina Arps-Forker
- Department of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Steven W Poser
- Department of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Feodor-Lynen Str. 25, 81377, Munich, Germany
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | | | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Feodor-Lynen Str. 25, 81377, Munich, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University Munich, EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Gregor-Mendel-Str. 2, 85350, Freising-Weihenstephan, Germany.,ZIEL - Institute for Food and Health, Technical University Munich, Gregor-Mendel-Str. 2, 85350, Freising-Weihenstephan, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Carsten Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technische Universität München, and Helmholtz Zentrum München, Ingolstädter Landstr.1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.,Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
| | - Vasiliki Tsata
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
| | | | - Maria Troullinaki
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Anke Witt
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Dresden, Germany
| | - Vivian Anastasiou
- DZD/Paul Langerhans Institute Dresden of Helmholtz Centre Munich, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - George Chrousos
- First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Aghia Sophia Children's Hospital, Athens, Greece
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Cristina García-Cáceres
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Matthias H Tschöp
- Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, 85764, Neuherberg, Germany.,Division of Metabolic Diseases, Technische Universität München, 80333, Munich, Germany
| | - Stefan R Bornstein
- Department of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Andreas Androutsellis-Theotokis
- Department of Medicine, Technische Universität Dresden, Dresden, Germany. .,DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany. .,Division of Cancer and Stem Cells, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
419
|
Rotermund C, Machetanz G, Fitzgerald JC. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front Endocrinol (Lausanne) 2018; 9:400. [PMID: 30072954 PMCID: PMC6060268 DOI: 10.3389/fendo.2018.00400] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/27/2018] [Indexed: 12/12/2022] Open
Abstract
The search for treatments for neurodegenerative diseases is a major concern in light of today's aging population and an increasing burden on individuals, families, and society. Although great advances have been made in the last decades to understand the underlying genetic and biological cause of these diseases, only some symptomatic treatments are available. Metformin has long since been used to treat Type 2 Diabetes and has been shown to be beneficial in several other conditions. Metformin is well-tested in vitro and in vivo and an approved compound that targets diverse pathways including mitochondrial energy production and insulin signaling. There is growing evidence for the benefits of metformin to counteract age-related diseases such as cancer, cardiovascular disease, and neurodegenerative diseases. We will discuss evidence showing that certain neurodegenerative diseases and diabetes are explicitly linked and that metformin along with other diabetes drugs can reduce neurological symptoms in some patients and reduce disease phenotypes in animal and cell models. An interesting therapeutic factor might be how metformin is able to balance survival and death signaling in cells through pathways that are commonly associated with neurodegenerative diseases. In healthy neurons, these overarching signals keep energy metabolism, oxidative stress, and proteostasis in check, avoiding the dysfunction and neuronal death that defines neurodegenerative disease. We will discuss the biological mechanisms involved and the relevance of neuronal vulnerability and potential difficulties for future trials and development of therapies.
Collapse
Affiliation(s)
| | - Gerrit Machetanz
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Julia C. Fitzgerald
- German Centre for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
420
|
Rotermund C, Machetanz G, Fitzgerald JC. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front Endocrinol (Lausanne) 2018; 9:400. [PMID: 30072954 DOI: 10.3389/fendo.2018.00400/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/27/2018] [Indexed: 05/25/2023] Open
Abstract
The search for treatments for neurodegenerative diseases is a major concern in light of today's aging population and an increasing burden on individuals, families, and society. Although great advances have been made in the last decades to understand the underlying genetic and biological cause of these diseases, only some symptomatic treatments are available. Metformin has long since been used to treat Type 2 Diabetes and has been shown to be beneficial in several other conditions. Metformin is well-tested in vitro and in vivo and an approved compound that targets diverse pathways including mitochondrial energy production and insulin signaling. There is growing evidence for the benefits of metformin to counteract age-related diseases such as cancer, cardiovascular disease, and neurodegenerative diseases. We will discuss evidence showing that certain neurodegenerative diseases and diabetes are explicitly linked and that metformin along with other diabetes drugs can reduce neurological symptoms in some patients and reduce disease phenotypes in animal and cell models. An interesting therapeutic factor might be how metformin is able to balance survival and death signaling in cells through pathways that are commonly associated with neurodegenerative diseases. In healthy neurons, these overarching signals keep energy metabolism, oxidative stress, and proteostasis in check, avoiding the dysfunction and neuronal death that defines neurodegenerative disease. We will discuss the biological mechanisms involved and the relevance of neuronal vulnerability and potential difficulties for future trials and development of therapies.
Collapse
Affiliation(s)
| | - Gerrit Machetanz
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Julia C Fitzgerald
- German Centre for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
421
|
Ursini F, Russo E, Pellino G, D'Angelo S, Chiaravalloti A, De Sarro G, Manfredini R, De Giorgio R. Metformin and Autoimmunity: A "New Deal" of an Old Drug. Front Immunol 2018; 9:1236. [PMID: 29915588 PMCID: PMC5994909 DOI: 10.3389/fimmu.2018.01236] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Metformin (dimethyl biguanide) is a synthetic derivative of guanidine, isolated from the extracts of Galega officinalis, a plant with a prominent antidiabetic effect. Since its discovery more than 50 years ago, metformin represents a worldwide milestone in treatment of patients with type 2 diabetes (T2D). Recent evidence in humans indicates novel pleiotropic actions of metformin which span from its consolidated role in T2D management up to various regulatory properties, including cardio- and nephro-protection, as well as antiproliferative, antifibrotic, and antioxidant effects. These findings, together with ground-breaking studies demonstrating its ability to prolong healthspan and lifespan in mice, provided the basis for defining metformin as a potential antiaging molecule. Moreover, emerging in vivo and in vitro evidence support the novel hypothesis that metformin can exhibit immune-modulatory features. Studies suggest that metformin interferes with key immunopathological mechanisms involved in systemic autoimmune diseases, such as the T helper 17/regulatory T cell balance, germinal centers formation, autoantibodies production, macrophage polarization, cytokine synthesis, neutrophil extracellular traps release, and bone or extracellular matrix remodeling. These effects may represent a powerful contributor to antiaging and anticancer properties exerted by metformin and, from another standpoint, may open the way to assess whether metformin can be a candidate molecule for clinical trials involving patients with immune-mediated diseases. In this article, we will review the available preclinical and clinical evidence regarding the effect of metformin on individual cells of the immune system, with emphasis on immunological mechanisms related to the development and maintenance of autoimmunity and its potential relevance in treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Gianluca Pellino
- Colorectal Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain.,Department of Medical, Surgical, Neurological, Metabolic and Ageing Sciences, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL) - Rheumatology Department of Lucania, "San Carlo" Hospital of Potenza and "Madonna delle Grazie" Hospital of Matera, Potenza, Italy.,Basilicata Ricerca Biomedica (BRB) Foundation, Potenza, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, Rome, Italy.,Department of Nuclear Medicine, IRCCS Neuromed, Pozzilli, Italy
| | | | - Roberto Manfredini
- Department of Medical Sciences, Clinica Medica Unit, University of Ferrara, Ferrara, Italy
| | - Roberto De Giorgio
- Department of Medical Sciences, Clinica Medica Unit, University of Ferrara, Ferrara, Italy
| |
Collapse
|
422
|
Varjabedian L, Bourji M, Pourafkari L, Nader ND. Cardioprotection by Metformin: Beneficial Effects Beyond Glucose Reduction. Am J Cardiovasc Drugs 2018; 18:181-193. [PMID: 29478240 DOI: 10.1007/s40256-018-0266-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metformin is a biguanide that is widely used as an insulin-sparing agent to treat diabetes. When compared with the general population, diabetics are twice as likely to die from fatal myocardial infarction and congestive heart failure (CHF). There has been a significant concern regarding the use of metformin in patients with CHF because of their higher tendency to develop lactic acidosis. However, large epidemiological trials have reported better cardiovascular prognosis with metformin compared to other glucose-lowering agents among diabetics. Additionally, metformin has reduced the risk of reinfarction and all-cause mortality in patients with coronary artery disease and CHF, respectively. The protection against cardiovascular diseases appears to be independent of the anti-hyperglycemic effects of metformin. These effects are mediated through an increase in 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and by increased phosphorylation of endothelial nitric oxide synthase (eNOS) in cardiomyocytes with an increased production of nitric oxide (NO). Metformin preconditions the heart against ischemia-reperfusion injury and may improve myocardial remodeling after an ischemic insult. The preponderance of evidence currently suggests that metformin is safe in patients with CHF, prompting the Food and Drug Administration to remove CHF as a contraindication from the package insert of all generic metformin preparations. In this narrative, along with a limited meta-analysis of available studies, we have reviewed the pleiotropic (non-glucose-lowering) effects of metformin that potentially contribute to its cardioprotective properties. Additionally, we have reviewed issues surrounding the safety of metformin in patients with cardiac diseases.
Collapse
Affiliation(s)
| | - Mohammad Bourji
- Department of Medicine, University at Buffalo, Buffalo, NY, 14203, USA
| | - Leili Pourafkari
- Department of Anesthesiology, University at Buffalo, 77 Goodell Street # 550, Buffalo, NY, 14203, USA
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, 77 Goodell Street # 550, Buffalo, NY, 14203, USA.
| |
Collapse
|
423
|
Al-Dwairi A, Alqudah M, Al-Shboul O, Alfaqih M, Alomari D. Metformin exerts anti-inflammatory effects on mouse colon smooth muscle cells in vitro. Exp Ther Med 2018; 16:985-992. [PMID: 30116349 DOI: 10.3892/etm.2018.6222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 04/20/2018] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic incurable condition characterized by relapsing inflammation of the gut. Intestinal smooth muscle cells (SMCs) are affected structurally and functionally during IBD due to excessive production of different inflammatory mediators. Metformin is a widely used antidiabetic agent known to exert several anti-inflammatory effects in different tissues independently from its hypoglycemic effect. The aim of the present study was to investigate the effect of metformin on expression and secretion of different cytokines and chemokines from mouse colon SMCs (CSMCs) following induction of inflammation with lipopolysaccharide (LPS) in vitro. CSMCs from male BALB/c mice were isolated and cultured in Dulbecco's modified Eagle's medium and treated with LPS (1 µg/ml) and 0, 5, 10 or 20 mM metformin for 24 h. Expression and secretion of tumor necrosis factor-α (TNF-α), interleukin-1α (IL-1α), macrophage colony stimulating factor (M-CSF), T cell activation gene-3 (TCA-3) and stromal cell-derived factor-1 (SDF-1) was evaluated by ELISA. LPS-treated CSMCs demonstrated significantly increased expression of TNF-α, IL-1α, M-CSF, TCA-3 and SDF-1 when compared with the control group (P<0.05). Co-treatment with metformin (5 and 10 mM) significantly reduced their expression by ~20-40% when compared with LPS treatment alone (P<0.05). Furthermore, secretion of TNF-α, IL-1α, M-CSF and TCA-3 into the conditioned media was significantly decreased by metformin (5 and 10 mM; P<0.05). In addition, metformin decreased levels of LPS-induced nuclear factor-κB phosphorylation. These data suggest that metformin may provide beneficial anti-inflammatory effects on CSMCs and it may be utilized as an adjunct therapy for patients suffering from IBD.
Collapse
Affiliation(s)
- Ahmed Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad Alqudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Othman Al-Shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mahmoud Alfaqih
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Dana Alomari
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
424
|
Packer M. Epicardial Adipose Tissue May Mediate Deleterious Effects of Obesity and Inflammation on the Myocardium. J Am Coll Cardiol 2018; 71:2360-2372. [PMID: 29773163 DOI: 10.1016/j.jacc.2018.03.509] [Citation(s) in RCA: 354] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/20/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023]
Abstract
Epicardial adipose tissue has unique properties that distinguish it from other depots of visceral fat. Rather than having distinct boundaries, the epicardium shares an unobstructed microcirculation with the underlying myocardium, and in healthy conditions, produces cytokines that nourish the heart. However, in chronic inflammatory disorders (especially those leading to heart failure with preserved ejection fraction), the epicardium becomes a site of deranged adipogenesis, leading to the secretion of proinflammatory adipokines that can cause atrial and ventricular fibrosis. Accordingly, in patients at risk of heart failure with preserved ejection fraction, drugs that promote the accumulation or inflammation of epicardial adipocytes may lead to heart failure, whereas treatments that ameliorate the proinflammatory characteristics of epicardial fat may reduce the risk of heart failure. These observations suggest that epicardial adipose tissue is a transducer of the adverse effects of systemic inflammation and metabolic disorders on the heart, and thus, represents an important target for therapeutic interventions.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas.
| |
Collapse
|
425
|
Malik F, Mehdi SF, Ali H, Patel P, Basharat A, Kumar A, Ashok F, Stein J, Brima W, Malhotra P, Roth J. Is metformin poised for a second career as an antimicrobial? Diabetes Metab Res Rev 2018; 34:e2975. [PMID: 29271563 DOI: 10.1002/dmrr.2975] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/24/2022]
Abstract
Metformin, a widely used antihyperglycaemic, has a good safety profile, reasonably manageable side-effects, is inexpensive, and causes a desirable amount of weight loss. In 4 studies of patients with tuberculosis (1 prospective and 3 retrospective), metformin administration resulted in better outcomes. In mice with several models of endotoxemia, metformin diminished levels of proinflammatory cytokines and improved survival. Laboratory studies showed effectiveness of the drug on multiple pathogens, including Trichinella spiralis, Staphylococcus aureus, Pseudomonas aeruginosa, hepatitis B virus, hepatitis C virus, and human immunodeficiency virus. Metformin administration in humans and mice produced major changes in the composition of the gut microbiota. These recently discovered microbe-modulating properties of the drug have led investigators to predict wide therapeutic utility for metformin. The recent easing in United States Food and Drug Administration (FDA) guidelines regarding administration of metformin to patients with kidney disease, and reduced anxiety about patient safety in terms of lactic acidosis, increase the probability of broadening of metformin's usage as a treatment of infectious agents. In this text we review articles pertinent to metformin's effects on microorganisms, both pathogens and commensals. We highlight the possible role of metformin in a wide range of infectious diseases and a possible expansion of its therapeutic profile in this field. A systematic review was done of PubMed indexed articles that examined the effects of metformin on a wide range of pathogens. Metformin was found to have efficacy as an antimicrobial agent in patients with tuberculosis. Mice infected with Trypanosomiasis cruzi had higher survival when also treated with metformin. The drug in vitro was active against T. spiralis, S. aureus, P. aeruginosa, and hepatitis B virus. In addition there is emerging literature on its role in sepsis. We conclude that metformin may have a potential role in the therapy for multiple infectious diseases. Metformin, in addition to its traditional effects on glucose metabolism, provides anti-microbial benefits in patients with tuberculosis and in a very wide range of other infections encounters in vitro and in vivo.
Collapse
Affiliation(s)
- Faiza Malik
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Syed Faizan Mehdi
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Haroon Ali
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Priya Patel
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Anam Basharat
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Amrat Kumar
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Fnu Ashok
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Joanna Stein
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Wunnie Brima
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Prashant Malhotra
- Division of Infectious Diseases, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Jesse Roth
- Laboratory of Diabetes and Diabetes-Related Research, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| |
Collapse
|
426
|
Cellular Senescence and Inflammaging in Age-Related Diseases. Mediators Inflamm 2018; 2018:9076485. [PMID: 29849499 PMCID: PMC5932453 DOI: 10.1155/2018/9076485] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/15/2018] [Indexed: 01/14/2023] Open
|
427
|
Kulkarni AS, Brutsaert EF, Anghel V, Zhang K, Bloomgarden N, Pollak M, Mar JC, Hawkins M, Crandall JP, Barzilai N. Metformin regulates metabolic and nonmetabolic pathways in skeletal muscle and subcutaneous adipose tissues of older adults. Aging Cell 2018; 17:e12723. [PMID: 29383869 PMCID: PMC5847877 DOI: 10.1111/acel.12723] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2017] [Indexed: 11/30/2022] Open
Abstract
Administration of metformin increases healthspan and lifespan in model systems, and evidence from clinical trials and observational studies suggests that metformin delays a variety of age-related morbidities. Although metformin has been shown to modulate multiple biological pathways at the cellular level, these pleiotropic effects of metformin on the biology of human aging have not been studied. We studied ~70-year-old participants (n = 14) in a randomized, double-blind, placebo-controlled, crossover trial in which they were treated with 6 weeks each of metformin and placebo. Following each treatment period, skeletal muscle and subcutaneous adipose tissue biopsies were obtained, and a mixed-meal challenge test was performed. As expected, metformin therapy lowered 2-hour glucose, insulin AUC, and insulin secretion compared to placebo. Using FDR<0.05, 647 genes were differentially expressed in muscle and 146 genes were differentially expressed in adipose tissue. Both metabolic and nonmetabolic pathways were significantly influenced, including pyruvate metabolism and DNA repair in muscle and PPAR and SREBP signaling, mitochondrial fatty acid oxidation, and collagen trimerization in adipose. While each tissue had a signature reflecting its own function, we identified a cascade of predictive upstream transcriptional regulators, including mTORC1, MYC, TNF, TGFß1, and miRNA-29b that may explain tissue-specific transcriptomic changes in response to metformin treatment. This study provides the first evidence that, in older adults, metformin has metabolic and nonmetabolic effects linked to aging. These data can inform the development of biomarkers for the effects of metformin, and potentially other drugs, on key aging pathways.
Collapse
Affiliation(s)
- Ameya S Kulkarni
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
- Institute for Clinical and Translational ResearchAlbert Einstein College of MedicineBronxNYUSA
| | - Erika F Brutsaert
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
| | - Valentin Anghel
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
| | - Kehao Zhang
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
| | - Noah Bloomgarden
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
| | | | - Jessica C Mar
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
- Department of Systems and Computational BiologyAlbert Einstein College of MedicineBronxNYUSA
- Australian Institute for Bioengineering and NanotechnologyUniversity of QueenslandBrisbaneQLDAustralia
| | - Meredith Hawkins
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
- Diabetes Research CenterAlbert Einstein College of MedicineBronxNYUSA
| | - Jill P Crandall
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
- Diabetes Research CenterAlbert Einstein College of MedicineBronxNYUSA
| | - Nir Barzilai
- Division of EndocrinologyDepartment of MedicineAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNYUSA
- Diabetes Research CenterAlbert Einstein College of MedicineBronxNYUSA
| |
Collapse
|
428
|
Cameron AR, Logie L, Patel K, Erhardt S, Bacon S, Middleton P, Harthill J, Forteath C, Coats JT, Kerr C, Curry H, Stewart D, Sakamoto K, Repiščák P, Paterson MJ, Hassinen I, McDougall G, Rena G. Metformin selectively targets redox control of complex I energy transduction. Redox Biol 2018; 14:187-197. [PMID: 28942196 PMCID: PMC5609876 DOI: 10.1016/j.redox.2017.08.018] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/15/2017] [Accepted: 08/25/2017] [Indexed: 11/24/2022] Open
Abstract
Many guanide-containing drugs are antihyperglycaemic but most exhibit toxicity, to the extent that only the biguanide metformin has enjoyed sustained clinical use. Here, we have isolated unique mitochondrial redox control properties of metformin that are likely to account for this difference. In primary hepatocytes and H4IIE hepatoma cells we found that antihyperglycaemic diguanides DG5-DG10 and the biguanide phenformin were up to 1000-fold more potent than metformin on cell signalling responses, gluconeogenic promoter expression and hepatocyte glucose production. Each drug inhibited cellular oxygen consumption similarly but there were marked differences in other respects. All diguanides and phenformin but not metformin inhibited NADH oxidation in submitochondrial particles, indicative of complex I inhibition, which also corresponded closely with dehydrogenase activity in living cells measured by WST-1. Consistent with these findings, in isolated mitochondria, DG8 but not metformin caused the NADH/NAD+ couple to become more reduced over time and mitochondrial deterioration ensued, suggesting direct inhibition of complex I and mitochondrial toxicity of DG8. In contrast, metformin exerted a selective oxidation of the mitochondrial NADH/NAD+ couple, without triggering mitochondrial deterioration. Together, our results suggest that metformin suppresses energy transduction by selectively inducing a state in complex I where redox and proton transfer domains are no longer efficiently coupled.
Collapse
Affiliation(s)
- Amy R Cameron
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Lisa Logie
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Kashyap Patel
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee, Scotland, UK
| | - Stefan Erhardt
- Institute of Chemical Sciences, School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, Scotland, UK
| | - Sandra Bacon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Paul Middleton
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Jean Harthill
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Calum Forteath
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Josh T Coats
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Calum Kerr
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Heather Curry
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK; Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Derek Stewart
- Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK; Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, Scotland, UK
| | - Kei Sakamoto
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dow Street, Dundee, Scotland, UK
| | - Peter Repiščák
- Institute of Chemical Sciences, School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, Scotland, UK
| | - Martin J Paterson
- Institute of Chemical Sciences, School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, Scotland, UK
| | - Ilmo Hassinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Gordon McDougall
- Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, Scotland, UK
| | - Graham Rena
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK.
| |
Collapse
|
429
|
Rakotoarivelo V, Lacraz G, Mayhue M, Brown C, Rottembourg D, Fradette J, Ilangumaran S, Menendez A, Langlois MF, Ramanathan S. Inflammatory Cytokine Profiles in Visceral and Subcutaneous Adipose Tissues of Obese Patients Undergoing Bariatric Surgery Reveal Lack of Correlation With Obesity or Diabetes. EBioMedicine 2018; 30:237-247. [PMID: 29548899 PMCID: PMC5952229 DOI: 10.1016/j.ebiom.2018.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023] Open
Abstract
Population studies have linked insulin resistance to systemic low-grade chronic inflammation and have reported elevated levels of inflammatory cytokines such as TNFα, IL-1β and IL-6, individually or in certain combinations, in adipose tissues or in the serum. We undertook this comprehensive study to simultaneously evaluate the expression of several pro-inflammatory and anti-inflammatory cytokines in serum and in the visceral and subcutaneous adipose tissues from obese patients undergoing bariatric surgery. We observed that several inflammatory cytokines implicated in obesity-associated inflammation showed no significant difference in protein or gene expression between obese patients with or without diabetes and control groups. IL1B gene expression was significantly elevated in the visceral adipose tissues of obese patients, but did not correlate with their diabetes status. Despite the significant increase in IL1B expression in the obese group, a significant proportion of obese patients did not express TNFA, IL1B or IL6 in visceral adipose tissues. Certain inflammatory cytokines showed correlation with the chemokine CCL2 and VEGF-A in visceral adipose tissues. Our findings suggest that the inflammatory cytokine profile in metabolic syndrome is more complex than what is currently perceived and that chronic inflammation in obese patients likely results from incremental contribution from different cytokines and possibly other inflammatory mediators from within and outside the adipose tissues. It is possible that this obesity associated chronic inflammation is not predicted by a single mediator, but rather includes a large spectrum of possible profiles. Visceral and subcutaneous adipose tissues do not express similar pattern of cytokines. VAT and SAT tissues from 30% of the obese patients do not express TNFA, IL6 or IL1B. Protein levels and gene expression do not necessarily correlate in VAT or SAT. The expression pattern of inflammatory mediators may present a larger spectrum than predicted from animal models.
Obesity, type 2 diabetes and cardiometabolic diseases are associated with a low-grade chronic inflammation. Various inflammatory mediators have been shown to mediate this inflammation. In this study we analyzed the expression of many of these inflammatory mediators in the visceral and subcutaneous adipose tissues obtained from patients undergoing bariatric surgery. Our results suggest that the profile of inflammatory mediators expressed in adipose tissue is diverse and varies from one patient to another.
Collapse
Affiliation(s)
- Volatiana Rakotoarivelo
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Gregory Lacraz
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marian Mayhue
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christine Brown
- Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Diane Rottembourg
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Division of Endocrinology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Julie Fradette
- Department of Surgery, Université laval, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Subburaj Ilangumaran
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Alfredo Menendez
- Department of Microbiology and Infectious diseases, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Marie-France Langlois
- Department of Medicine, Division of Endocrinology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Sheela Ramanathan
- Department of Pediatrics, Division of Immunology, Faculty of Medicine and Heath Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche, Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada.
| |
Collapse
|
430
|
Bishwakarma R, Zhang W, Lin YL, Kuo YF, Cardenas VJ, Sharma G. Metformin use and health care utilization in patients with coexisting chronic obstructive pulmonary disease and diabetes mellitus. Int J Chron Obstruct Pulmon Dis 2018; 13:793-800. [PMID: 29551895 PMCID: PMC5842767 DOI: 10.2147/copd.s150047] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is associated with persistent systemic inflammation. Anti-inflammatory therapies have been shown to decrease acute exacerbations of COPD. The antidiabetic medication metformin decreases oxidative stress and inflammation and may benefit patients with COPD. We aimed at investigating the effect of metformin on health care utilizations in patients with coexisting COPD and diabetes mellitus (DM). Methods We studied 5% Medicare beneficiaries with coexisting COPD and DM prescribed metformin or other antidiabetics during the period 2007-2010. The primary outcome was COPD-specific emergency room (ER) visits and hospitalizations; the secondary outcome was all-cause ER visits and hospitalizations over the 2-year follow-up after the index antidiabetic prescription. The effects of metformin were examined by COPD complexity and compared with the effects of other antidiabetic medications. Results Among 11,260 patients, 3,193 were metformin users and 8,067 were nonusers. Metformin users were younger, were less sick, were less likely to be on oxygen, and had fewer hospitalizations in the prior year compared with the nonusers. Over a 2-year period, metformin users had lower COPD-specific and all-cause ER visits and hospitalizations (7.11% vs 9.61%, p<0.0001; and 61.63% vs 71.27%, p<0.0001, respectively). In a stratified multivariable analysis, the odds of COPD-specific ER visits and hospitalizations were lower in patients with low-complexity COPD (adjusted odds ratio =0.66, 95% confidence interval =0.52-0.85). However, patients with all COPD complexities get benefits of metformin on all-cause ER visits and hospitalizations. Conclusion The use of metformin in patients with coexisting COPD and DM was associated with fewer COPD-specific ER visits and hospitalizations, especially in low-complexity COPD.
Collapse
Affiliation(s)
- Raju Bishwakarma
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
| | - Wei Zhang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
| | | | - Yong-Fang Kuo
- Office of Biostatistics
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX, USA
| | - Victor J Cardenas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
| | - Gulshan Sharma
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
431
|
Cardiac Autonomic Neuropathy as a Result of Mild Hypercaloric Challenge in Absence of Signs of Diabetes: Modulation by Antidiabetic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9389784. [PMID: 29643979 PMCID: PMC5831709 DOI: 10.1155/2018/9389784] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022]
Abstract
Cardiac autonomic neuropathy (CAN) is an early cardiovascular complication of diabetes occurring before metabolic derangement is evident. The cause of CAN remains elusive and cannot be directly linked to hyperglycemia. Recent clinical data report cardioprotective effects of some antidiabetic drugs independent of their hypoglycemic action. Here, we used a rat model receiving limited daily increase in calories from fat (HC diet) to assess whether mild metabolic challenge led to CAN in absence of interfering effects of hyperglycemia, glucose intolerance, or obesity. Rats receiving HC diet for 12 weeks showed reduction in baroreceptor sensitivity and heart rate variability despite lack of change in baseline hemodynamic and cardiovascular structural parameters. Impairment of cardiac autonomic control was accompanied with perivascular adipose inflammation observed as an increased inflammatory cytokine expression, together with increased cardiac oxidative stress, and signaling derangement characteristic of diabetic cardiomyopathy. Two-week treatment with metformin or pioglitazone rectified the autonomic derangement and corrected the molecular changes. Switching rats to normal chow but not to isocaloric amounts of HC for two weeks reversed CAN. As such, we conclude that adipose inflammation due to increased fat intake might underlie development of CAN and, hence, the beneficial effects of metformin and pioglitazone.
Collapse
|
432
|
Affiliation(s)
- Graham Rena
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Scotland, United Kingdom.
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Scotland, United Kingdom.
| |
Collapse
|
433
|
Huangfu N, Wang Y, Cheng J, Xu Z, Wang S. Metformin protects against oxidized low density lipoprotein-induced macrophage apoptosis and inhibits lipid uptake. Exp Ther Med 2018; 15:2485-2491. [PMID: 29456653 PMCID: PMC5795518 DOI: 10.3892/etm.2018.5704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/28/2017] [Indexed: 12/31/2022] Open
Abstract
Oxidized low density lipoprotein (ox-LDL)-induced macrophage apoptosis contributes to the formation of atherosclerosis. Metformin, an antidiabetic drug, has been reported to attenuate lipid accumulation in macrophages. In this study, the effects of metformin on ox-LDL-induced macrophage apoptosis were investigated and the mechanisms involved in this process were examined. By performing flow cytometry analysis, it was demonstrated that metformin inhibited ox-LDL-induced macrophage apoptosis. Increased expression of endoplasmic reticulum (ER) stress marker proteins, including C/EBP-homologous protein, eukaryotic translation initiation factor 2A, and glucose-regulated protein 78 kDa, induced by ox-LDL was also reversed by metformin. Furthermore, ox-LDL-induced cytochrome c (cyto-c) release and mitochondrial membrane potential loss were inhibited by metformin. As lipid uptake in macrophages contributed to ER stress, cyto-c release and mitochondrial membrane potential loss, the mechanisms involved in metformin-inhibited macrophage lipid uptake were investigated. Expression of scavenger receptors, including scavenger receptor A, cluster of differentiation 36 and lectin-type oxidized LDL receptor 1 was examined in the presence or absence of metformin with ox-LDL treatment. Additionally, the upstream regulatory mechanism of scavenger receptors by metformin was also analyzed. In conclusion, metformin protects against ox-LDL-induced macrophage apoptosis and inhibits macrophage lipid uptake.
Collapse
Affiliation(s)
- Ning Huangfu
- Department of Cardiology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Yong Wang
- Department of Cardiology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Jingsong Cheng
- Department of Cardiology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Zhenyu Xu
- Department of Cardiology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Shenghuang Wang
- Department of Cardiology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
434
|
Prattichizzo F, De Nigris V, Spiga R, Mancuso E, La Sala L, Antonicelli R, Testa R, Procopio AD, Olivieri F, Ceriello A. Inflammageing and metaflammation: The yin and yang of type 2 diabetes. Ageing Res Rev 2018; 41:1-17. [PMID: 29081381 DOI: 10.1016/j.arr.2017.10.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/21/2017] [Accepted: 10/23/2017] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is characterised by chronic low-grade inflammation, recently referred to as 'metaflammation', a relevant factor contributing to the development of both diabetes and its complications. Nonetheless, 'canonical' anti-inflammatory drugs do not yield satisfactory results in terms of prevention of diabetes progression and of cardiovascular events, suggesting that the causal mechanisms fostering metaflammation deserve further research to identify new druggable targets. Metaflammation resembles ageing-induced low-grade inflammation, previously referred to as inflammageing, in terms of clinical presentation and the molecular profile, pointing to a common aetiology for both conditions. Along with the mechanisms proposed to fuel inflammageing, here we dissect a plethora of pathological cascades triggered by gluco- and lipotoxicity, converging on candidate phenomena possibly explaining the enduring pro-inflammatory program observed in diabetic tissues, i.e. persistent immune-system stimulation, accumulation of senescent cells, epigenetic rearrangements, and alterations in microbiota composition. We discuss the possibility of harnessing these recent discoveries in future therapies for T2DM. Moreover, we review recent evidence regarding the ability of diets and physical exercise to modulate selected inflammatory pathways relevant for the diabetic pathology. Finally, we examine the latest findings showing putative anti-inflammatory mechanisms of anti-hyperglycaemic agents with proven efficacy against T2DM-induced cardiovascular complications, in order to gain insights into quickly translatable therapeutic approaches.
Collapse
|
435
|
Sosale B, Chandrashekara S, Aravind SR, Renuka P, Anupama K. Influence of cytokine status on insulin resistance and circulating endothelial progenitor cells in type 2 diabetes mellitus. Cytokine 2017; 99:179-185. [DOI: 10.1016/j.cyto.2017.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/30/2017] [Accepted: 09/10/2017] [Indexed: 12/27/2022]
|
436
|
Metformin therapy and postoperative atrial fibrillation in diabetic patients after cardiac surgery. J Intensive Care 2017; 5:60. [PMID: 29075499 PMCID: PMC5648492 DOI: 10.1186/s40560-017-0254-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/10/2017] [Indexed: 11/28/2022] Open
Abstract
Background Postoperative atrial fibrillation (AF) commonly occurs in cardiac surgery patients. Studies suggest inflammation and oxidative stress contribute to postoperative AF development in this patient population. Metformin exerts an anti-inflammatory effect that reduces oxidative stress and thus may play a role in preventing postoperative AF. Methods We conducted a matched, retrospective cohort study of diabetic patients’ age ≥18 undergoing a coronary artery bypass graft (CABG) and/or cardiac valve surgery from January 1, 2009, to November 30, 2014. We extracted data from The Society of Thoracic Surgeons National Adult Cardiac Surgery Database. Primary exposure was ongoing metformin use at a dose of ≥ 500 mg in effect before cardiac surgery as captured before admission. Primary study outcome was postoperative AF incidence. Matching was used to reduce selection bias between metformin and non-metformin groups. Comparison between the groups after matching was accomplished using the McNemar test or paired t test. Results Out of the 4177 patients with cardiac surgery (CABG and/or valve surgery), 1283 patients met our study criteria. These patients were grouped into metformin [n = 635 (49.5%)] and non-metformin [n = 648 (50.5%)] users. Pre-matching, postoperative AF was found in 149 (23.5%) patients in the metformin group and 172 (26.5%) in the non-metformin group (p = 0.2088). Matching resulted in a total of 114 patients in each group (metformin vs. non-metformin). We found no statistically significant difference for postoperative AF between the two groups after matching (p = 0.8964). Conclusions Prior use of metformin therapy in diabetic patients undergoing cardiac surgery was not associated with decreased rate of postoperative AF.
Collapse
|
437
|
Khajuria DK, Patil ON, Karasik D, Razdan R. Development and evaluation of novel biodegradable chitosan based metformin intrapocket dental film for the management of periodontitis and alveolar bone loss in a rat model. Arch Oral Biol 2017; 85:120-129. [PMID: 29055230 DOI: 10.1016/j.archoralbio.2017.10.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of this study was to develop a chitosan-metformin based intrapocket dental film (CMIDF) for applications in the treatment of periodontitis and alveolar bone loss in an rat model of periodontitis. DESIGN CMIDF inserts were fabricated by the solvent casting technique. The fabricated inserts were evaluated for physical characteristics such as folding endurance, surface pH, mucoadhesive strength, metformin content uniformity, and release. X-ray diffraction analysis indicates no crystallinity of metformin in presence of chitosan which confirmed successful entrapment of metformin into the CMIDF. Fourier-transform infrared spectroscopy revealed stability of CMIDF and compatibility between metformin and chitosan. Periodontitis was induced by a combination of Porphyromonas gingivalis- lipopolysaccharide injections in combinations with ligatures around the mandibular first molar. We divided rats into 5 groups (8 rats/group): healthy, untreated periodontitis; periodontitis plus CMIDF-A (1.99±0.09mg metformin; total mass-4.01±0.05mg), periodontitis plus CMIDF-B (2.07±0.06mg metformin; total mass-7.56±0.09mg), and periodontitis plus chitosan film (7.61±0.08mg). After four weeks, mandibles were extracted to evaluate alveolar bone loss by micro-computerized tomography and histological techniques. RESULTS Alveolar bone was intact in the healthy group. Local administration of CMIDF resulted in significant improvements in the alveolar bone properties when compared to the untreated periodontitis group. The study reported here demonstrates that novel CMIDF showed good antibacterial activity and effectively reduced alveolar bone destruction in a rat model of experimental periodontitis. CONCLUSIONS Novel CMIDF showed good antibacterial activity and improved alveolar bone properties in a rat model.
Collapse
Affiliation(s)
- Deepak Kumar Khajuria
- The Musculoskeletal Genetics Laboratory, Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel; Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, India.
| | | | - David Karasik
- The Musculoskeletal Genetics Laboratory, Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Rema Razdan
- Department of Pharmacology, Al-Ameen College of Pharmacy, Bangalore, India
| |
Collapse
|
438
|
Zheng C, Hu X, Zhao L, Hu M, Gao F. Clinical and pharmacological hallmarks of rifapentine's use in diabetes patients with active and latent tuberculosis: do we know enough? DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2957-2968. [PMID: 29066867 PMCID: PMC5644564 DOI: 10.2147/dddt.s146506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Rifapentine is a rifamycin derivate approved by the US Food and Drug Administration in 1998 for the treatment of active, drug-susceptible tuberculosis (TB). In 2014, rifapentine was approved for the treatment of latent TB infection in patients at high risk of progression to active disease and is currently under evaluation by the European Medicines Agency. Expanding indications of rifapentine largely affect diabetes patients, since about one-third of them harbor latent TB. Clinical consequences of rifapentine use in this population and potentially harmful interactions with hypoglycemic agents are widely underexplored and generally considered similar to the ones of rifampicin. Indeed, rifapentine too may decrease blood levels of many oral antidiabetics and compete with them for protein-binding sites and/or transporters. However, the two drugs differ in protein-binding degree, the magnitude of cytochrome P450 induction and auto-induction, the degree of renal elimination, and so on. Rifapentine seems to be more suitable for use in diabetes patients with renal impairment, owing to the fact that it does not cause renal toxicity, and it is eliminated via kidneys in smaller proportions than rifampicin. On the other hand, there are no data related to rifapentine use in patients >65 years, and hypoalbuminemia associated with diabetic kidney disease may affect a free fraction of rifapentine to a greater extent than that of rifampicin. Until more pharmacokinetic information and information on the safety of rifapentine use in diabetic patients and drug–drug interactions are available, diabetes in TB patients treated with rifapentine should be managed with insulin analogs, and glucose and rifapentine plasma levels should be closely monitored.
Collapse
Affiliation(s)
- Chunlan Zheng
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Xiufen Hu
- Department of Paediatrics, Tongji Hospital
| | - Li Zhao
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Minhui Hu
- Department of Internal Medicine - Section 5, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Control Institute)
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
439
|
AMPK is not required for the effect of metformin on the inhibition of BMP6-induced hepcidin gene expression in hepatocytes. Sci Rep 2017; 7:12679. [PMID: 28978947 PMCID: PMC5627262 DOI: 10.1038/s41598-017-12976-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 09/13/2017] [Indexed: 12/23/2022] Open
Abstract
The biguanide metformin is used for its antidiabetic effect for many years but how metformin acts remains poorly understood and controversial. AMP-activated protein kinase (AMPK), a protein kinase that plays a key role in maintaining energy homeostasis, is assumed to be one of the prime targets of metformin. However, since our demonstration that AMPK is not required for the beneficial effects of metformin on the control of glycemia, the list of AMPK-independent actions of metformin is rapidly increasing. Given the conflicting results on the effects of metformin we sought, using our genetic mouse models deficient in the catalytic subunits of AMPK, to determine whether this kinase is involved in the effects of metformin on the expression of the iron-regulatory hormone hepcidin, as recently proposed. Here we demonstrate, using different approaches, either isolated hepatocytes that lack AMPK, or direct AMPK activators, that, AMPK activation is not necessary for metformin to inhibit BMP6-induced hepcidin gene expression. These results may shed new lights on the increasingly recognized AMPK-independent metformin's molecular action, an important area of current research.
Collapse
|
440
|
Frati G, Schirone L, Chimenti I, Yee D, Biondi-Zoccai G, Volpe M, Sciarretta S. An overview of the inflammatory signalling mechanisms in the myocardium underlying the development of diabetic cardiomyopathy. Cardiovasc Res 2017; 113:378-388. [PMID: 28395009 DOI: 10.1093/cvr/cvx011] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/20/2017] [Indexed: 02/05/2023] Open
Abstract
Heart failure is a highly morbid and mortal clinical condition that represents the last stage of most cardiovascular disorders. Diabetes is strongly associated with an increased incidence of heart failure and directly promotes cardiac hypertrophy, fibrosis, and apoptosis. These changes, in turn, contribute to the development of ventricular dysfunction. The clinical condition associated with the spectrum of cardiac abnormalities induced by diabetes is termed diabetic cardiomyopathy. Myocardial inflammation has recently emerged as a pathophysiological process contributing to cardiac hypertrophy, fibrosis, and dysfunction in cardiac diseases. Myocardial inflammation is also implicated in the development of diabetic cardiomyopathy. Several molecular mechanisms link diabetes to myocardial inflammation. The NF-κB signalling pathway and the renin-angiotensin-aldosterone system are strongly activated in the diabetic heart, thereby promoting myocardial inflammation. Advanced glycation end-products and damage-associated molecular pattern molecules also represent strong triggers for inflammation. The mediators resulting from this inflammatory process modulate specific intracellular signalling mechanisms in cardiac cells that promote the development of diabetic cardiomyopathy. This review article will provide an overview of the signalling molecular mechanisms linking diabetic cardiomyopathy to myocardial inflammation.
Collapse
Affiliation(s)
- Giacomo Frati
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy
| | - Isotta Chimenti
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy
| | - Derek Yee
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Giuseppe Biondi-Zoccai
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| | - Massimo Volpe
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, 04100 Latina (LT), Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| |
Collapse
|
441
|
Uddin R, Nur-E-Habiba, Rena G, Hwu ET, Boisen A. New Evidence for the Mechanism of Action of a Type-2 Diabetes Drug Using a Magnetic Bead-Based Automated Biosensing Platform. ACS Sens 2017; 2:1329-1336. [PMID: 28776376 PMCID: PMC5613276 DOI: 10.1021/acssensors.7b00384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
![]()
The mechanism of
action (MOA) of the first line type-2 diabetes
drug metformin remains unclear despite its widespread usage. However,
recent evidence suggests that the mitochondrial copper (Cu)-binding
action of metformin may contribute toward the drug’s MOA. Here,
we present a novel biosensing platform for investigating the MOA of
metformin using a magnetic microbead-based agglutination assay which
has allowed us to demonstrate for the first time the interaction between
Cu and metformin at clinically relevant low micromolar concentrations
of the drug, thus suggesting a potential pathway of metformin’s
blood-glucose lowering action. In this assay, cysteine-functionalized
magnetic beadswere agglutinated in the presence of Cu due to cysteine’s
Cu-chelation property. Addition of clinically relevant doses of metformin
resulted in disaggregation of Cu-bridged bead-clusters, whereas the
effect of adding a closely related but blood-glucose neutral drug
propanediimidamide (PDI) showed completely different responses to
the clusters. The entire assay was integrated in an automated microfluidics
platform with an advanced optical imaging unit by which we investigated
these aggregation–disaggregation phenomena in a reliable, automated,
and user-friendly fashion with total assay time of 17 min requiring
a sample (metformin/PDI) volume of 30 μL. The marked difference
of Cu-binding action between the blood-glucose lowering drug metformin
and its inactive analogue PDI thus suggests that metformin’s
distinctive Cu-binding properties may be required for its effect on
glucose homeostasis. The novel automated platform demonstrating this
novel investigation thus holds the potential to be utilized for investigating
significant and sensitive molecular interactions via magnetic bead-based
agglutination assay.
Collapse
Affiliation(s)
- Rokon Uddin
- Department
of Micro- and Nanotechnology, Technical University of Denmark, DTU Nanotech, Building 345 East, DK-2800 Kongens Lyngby, Denmark
| | - Nur-E-Habiba
- Department
of Chemistry and Bioengineering, Tampere University of Technology, 33720, Tampere, Finland
| | - Graham Rena
- Molecular
and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - En-Te Hwu
- Institute
of Physics, Academia Sinica, Nankang, 11529 Taiwan
| | - Anja Boisen
- Department
of Micro- and Nanotechnology, Technical University of Denmark, DTU Nanotech, Building 345 East, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|
442
|
Patrice NK, Martial DJ, Bruno TP. Is it a supplementary benefit to use anti-inflammatory agents in the treatment of type 2 diabetes? BMC Res Notes 2017; 10:471. [PMID: 28886725 PMCID: PMC5591512 DOI: 10.1186/s13104-017-2785-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate an independent correlation between high sensitivity C-reactive protein (hs-CRP) and glycated haemoglobin (HbA1c) on one side and between hs-CRP and arterial pressure in well glucose controlled type 2 diabetics on the other side. RESULTS The mean of HbA1c was 6.37% in this study and 70.10% of participants had an HbA1c less than 7%. A positive correlation between hs-CRP and HbA1c was found in the current study (r = 0.232; P = 0.043) and we observed a decrease of 0.79% of HbA1c of the participants when their hs-CRP was less than 1 mg/l compared to that of the participants who had hs-CRP more than 1 mg/l (5.75 ± 1.25% VS 6.54 ± 1.42% P value = 0.04 Student). No correlation was found between hs-CRP and arterial pressure in this study. These results could justify the perspective use of anti-inflammatory drugs in the management of T2D. However the presence of participants with HbA1c levels greater than 7% makes plausible the influence of confounding factors on the observed correlations.
Collapse
Affiliation(s)
- Nzogang K. Patrice
- Department of Biochemistry, University of Dschang and District Hospital of Batcham (West Cameroon), Dschang, Cameroon
| | - Donkeng J. Martial
- Department of Biochemistry, University of Dschang and District Hospital of Tibati (Adamaoua Cameroon), Dschang, Cameroon
| | - Telefo P. Bruno
- Department of Biochemistry, University of Dschang, Dschang, Cameroon
| |
Collapse
|
443
|
Heckman-Stoddard BM, DeCensi A, Sahasrabuddhe VV, Ford LG. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia 2017; 60:1639-1647. [PMID: 28776080 PMCID: PMC5709147 DOI: 10.1007/s00125-017-4372-6] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 01/16/2023]
Abstract
Multiple epidemiological studies have documented an association between metformin, used for treatment of type 2 diabetes, and reduced cancer incidence and mortality. Cell line models may not accurately reflect the effects of metformin in the clinical setting. Moreover, findings from animal model studies have been inconsistent, whilst those from more recent epidemiological studies have tempered the overall effect size. The purpose of this review is to examine metformin's chemopreventive potential by outlining relevant mechanisms of action, the most recent epidemiologic evidence, and recently completed and ongoing clinical trials. Although repurposing drugs with excellent safety profiles is an appealing strategy for cancer prevention and treatment in the adjuvant setting, there is no substitute for well-executed, large randomised clinical trials to define efficacy and determine the populations that are most likely to benefit from an intervention. Thus, enthusiasm remains for understanding the role of metformin in cancer through ongoing clinical research.
Collapse
Affiliation(s)
- Brandy M Heckman-Stoddard
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA.
| | - Andrea DeCensi
- Division of Medical Oncology, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Vikrant V Sahasrabuddhe
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Leslie G Ford
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
444
|
Abstract
Metformin is a widely-used drug that results in clear benefits in relation to glucose metabolism and diabetes-related complications. The mechanisms underlying these benefits are complex and still not fully understood. Physiologically, metformin has been shown to reduce hepatic glucose production, yet not all of its effects can be explained by this mechanism and there is increasing evidence of a key role for the gut. At the molecular level the findings vary depending on the doses of metformin used and duration of treatment, with clear differences between acute and chronic administration. Metformin has been shown to act via both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms; by inhibition of mitochondrial respiration but also perhaps by inhibition of mitochondrial glycerophosphate dehydrogenase, and a mechanism involving the lysosome. In the last 10 years, we have moved from a simple picture, that metformin improves glycaemia by acting on the liver via AMPK activation, to a much more complex picture reflecting its multiple modes of action. More work is required to truly understand how this drug works in its target population: individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Graham Rena
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Ewan R Pearson
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
445
|
Freckelton J, Evans JA, Croagh D, Moore GT. Metformin use in diabetics with diverticular disease is associated with reduced incidence of diverticulitis. Scand J Gastroenterol 2017; 52:969-972. [PMID: 28503973 DOI: 10.1080/00365521.2017.1325930] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS There is no current, evidence-based therapy to prevent acute diverticulitis in patients with diverticular disease. Metformin has been shown to have anti-inflammatory effects in a number of disease states, in both animal models and in human observational studies. The potential therapeutic efficacy of metformin in diverticular disease has not been investigated. This study aims to describe the relationship between metformin use and diverticular disease in patients with diabetes mellitus. METHODS This was a retrospective case-control study. It compared metformin and other hypoglycaemic medication use in diabetic patients with uncomplicated diverticulosis to those with acute diverticulitis. Patients were identified using hospital International Classification of Diseases 10 (ICD-10) data, and radiology, pathology and scanned medical record databases were used to confirm diagnoses and collect all information. Chi square tests were used to determine significance of difference in categorical variables, and Mann-Whitney tests were used for continuous data. RESULTS There were 174 patients with uncomplicated diverticulosis and 175 patients with acute diverticulitis. A diagnosis of acute diverticulitis was associated with a significantly lower incidence of metformin use, than a diagnosis of uncomplicated diverticular disease (44% compared to 60%, respectively, p = .002). Other oral hypoglycaemic drugs and insulin were not associated with a lower incidence of diverticulitis (p = .12 and p = .24, respectively). CONCLUSION Metformin use is associated with reduced incidence of diverticulitis in diabetic patients with diverticular disease. The utility of metformin as a therapeutic agent to reduce the risk of diverticulitis in patients with diverticular disease warrants further randomised, prospective, interventional investigation.
Collapse
Affiliation(s)
- Julia Freckelton
- a Department of Medicine, School of Clinical Sciences at Monash Health , Monash University , Clayton , Australia
| | - Jacqueline Amanda Evans
- a Department of Medicine, School of Clinical Sciences at Monash Health , Monash University , Clayton , Australia
| | - Daniel Croagh
- b Department of Surgery, School of Clinical Sciences at Monash Health , Monash University , Clayton , Australia
| | - Gregory Thomas Moore
- c Centre for Inflammatory Diseases , School of Clinical Sciences at Monash Health, Monash University , Clayton , Australia
| |
Collapse
|
446
|
Pollak M. The effects of metformin on gut microbiota and the immune system as research frontiers. Diabetologia 2017; 60:1662-1667. [PMID: 28770326 DOI: 10.1007/s00125-017-4352-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/15/2017] [Indexed: 12/21/2022]
Abstract
Recent studies have revealed that metformin influences gut microbiota and the immune system although neither is a classic target of the drug. This research has revealed complexity not previously appreciated, and opened new research directions. The extent to which immunomodulatory effects and actions on the microbiota are related to each other and account for effects on host energy metabolism remains to be determined. These sites of action may be relevant not only to the efficacy of metformin for its established use in type 2 diabetes, but also to proposed novel indications in oncology and other diseases.
Collapse
Affiliation(s)
- Michael Pollak
- Division of Cancer Prevention, Department of Oncology, McGill University, Montreal, QC, H3T 1E2, Canada.
| |
Collapse
|
447
|
Chang MY, Ma TL, Hung CC, Tian YC, Chen YC, Yang CW, Cheng YC. Metformin Inhibits Cyst Formation in a Zebrafish Model of Polycystin-2 Deficiency. Sci Rep 2017; 7:7161. [PMID: 28769124 PMCID: PMC5541071 DOI: 10.1038/s41598-017-07300-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/26/2017] [Indexed: 12/27/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common kidney disease caused by mutations in PKD1 or PKD2. Metformin reduces cyst growth in mouse models of PKD1. However, metformin has not been studied in animal models of PKD2, and the cellular mechanism underlying its effectiveness is not entirely clear. This study investigated the effects of metformin on cyst formation in a zebrafish model of polycystin-2 deficiency resulting from morpholino knockdown of pkd2. We added metformin (2.5 to 20 mM) to the embryo media between 4 and 48 hours post fertilisation and observed pronephric cyst formation by using the wt1b promoter-driven GFP signal in Tg(wt1b:GFP) pkd2 morphants. Metformin inhibited pronephric cyst formation by 42–61% compared with the untreated controls. Metformin also reduced the number of proliferating cells in the pronephric ducts, the degree of dorsal body curvature, and the infiltration of leukocytes surrounding the pronephros. Moreover, metformin treatment increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and enhanced autophagy in the pronephros. Our data suggest that metformin reduces cyst formation through activation of the AMPK pathway and modulation of defective cellular events such as proliferation and autophagy. These results also imply that metformin could have therapeutic potential for ADPKD treatment.
Collapse
Affiliation(s)
- Ming-Yang Chang
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tsu-Lin Ma
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Cheng-Chieh Hung
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ya-Chung Tian
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Chang Chen
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Wei Yang
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.
| |
Collapse
|
448
|
Abstract
The AMP-activated protein kinase (AMPK) is a central regulator of multiple metabolic pathways and may have therapeutic importance for treating obesity, insulin resistance, type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), and cardiovascular disease (CVD). Given the ubiquitous expression of AMPK, it has been a challenge to evaluate which tissue types may be most beneficially poised for mediating the positive metabolic effects of AMPK-centered treatments. In this review we evaluate the metabolic phenotypes of transgenic mouse models in which AMPK expression and function have been manipulated, and the impact this has on controlling lipid metabolism, glucose homeostasis, and inflammation. This information may be useful for guiding the development of AMPK-targeted therapeutics to treat chronic metabolic diseases.
Collapse
Affiliation(s)
- Emily A Day
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Rebecca J Ford
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Gregory R Steinberg
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, Canada.
| |
Collapse
|
449
|
Sun X, Zhu MJ. AMP-activated protein kinase: a therapeutic target in intestinal diseases. Open Biol 2017; 7:170104. [PMID: 28835570 PMCID: PMC5577448 DOI: 10.1098/rsob.170104] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK), a highly conserved energy sensor, has a crucial role in cardiovascular, neurodegenerative and inflammatory diseases, as well as in cancer and metabolic disorders. Accumulating studies have demonstrated that AMPK activation enhances paracellular junctions, nutrient transporters, autophagy and apoptosis, and suppresses inflammation and carcinogenesis in the intestine, indicating an essential role of AMPK in intestinal health. AMPK inactivation is an aetiological factor in intestinal dysfunctions. This review summarizes the favourable outcomes of AMPK activation on intestinal health, and discusses AMPK as a potential therapeutic target for intestinal diseases.
Collapse
Affiliation(s)
- Xiaofei Sun
- School of Food Science, Washington State University, Pullman, WA 99164, USA
- School of Food Science, University of Idaho, Moscow, ID 83844, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA
- School of Food Science, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
450
|
Oliveira VC, Constante SAR, Orsolin PC, Nepomuceno JC, de Rezende AAA, Spanó MA. Modulatory effects of metformin on mutagenicity and epithelial tumor incidence in doxorubicin-treated Drosophila melanogaster. Food Chem Toxicol 2017; 106:283-291. [DOI: 10.1016/j.fct.2017.05.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/10/2017] [Accepted: 05/26/2017] [Indexed: 12/13/2022]
|