1
|
Sharrack N, Knott KD, Gulsin GS, Kotecha T, Brown LAE, Yeo JL, Porcari A, Adam RD, Thirunavukarasu S, Chowdhary A, Levelt E, Moon JC, McCann GP, Fontana M, Kellman P, Munyombwe T, Gale CP, Buckley DL, Greenwood JP, Swoboda PP, Plein S. Metformin associates with higher myocardial perfusion reserve and survival in type 2 diabetes mellitus patients. Sci Rep 2024; 14:27280. [PMID: 39516499 PMCID: PMC11549305 DOI: 10.1038/s41598-024-77280-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Metformin is an antihyperglycemic used to treat type 2 diabetes mellitus (T2DM). Patients with T2DM are at increased risk of cardiovascular disease. We explored the association between metformin use and cardiovascular magnetic resonance (CMR) derived stress myocardial blood flow (MBF), myocardial perfusion reserve (MPR) and major adverse cardiovascular events (MACE; all cause death, MI, stroke, heart failure hospitalisation and coronary revascularisation) in patients with T2DM. Multi-centre study of patients with T2DM, and healthy controls, underwent quantitative myocardial perfusion CMR using an artificial intelligence supported process. Multivariable regression analysis, and cox proportional hazard models of propensity score weighted patients quantified associations between metformin use, MBF, MPR, all cause death and MACE. Analysis included 572 patients with T2DM (68% prescribed metformin) with median follow-up 851 days (IQR 935 - 765). Metformin use was associated with an increase of MPR of 0.12 [0.08-0.40], p = 0.004. There were 82 MACE events (14.3%) including 25 (4.4%) deaths of which 16 were in those not prescribed metformin (8.7%), compared to 9 in patients prescribed metformin (2.3%): adjusted hazard ratio 0.24 (95% CI 0.08-0.70, p = 0.009). MACE events were similar between groups. This multicentre, inverse probability weighting propensity score analysis study showed that in patients with T2DM, metformin use is associated with higher MPR and improved all cause survival.
Collapse
Affiliation(s)
- Noor Sharrack
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| | - Kristopher D Knott
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Tushar Kotecha
- Institute of Cardiovascular Science, University College London, London, UK
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
| | - Louise A E Brown
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Jian L Yeo
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Aldostefano Porcari
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
- Centre for Diagnosis and Treatment of Cardiomyopathies, Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI), University of Trieste, Trieste, Italy
| | - Robert D Adam
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Sharmaine Thirunavukarasu
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Amrit Chowdhary
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Eylem Levelt
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - James C Moon
- Barts Heart Centre, St Bartholomew's Hospital, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, Cardiovascular Biomedical Research Centre, University of Leicester and the NIHR Leicester, University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, UK
| | - Marianna Fontana
- Institute of Cardiovascular Science, University College London, London, UK
- Division of Medicine, National Amyloidosis Centre, University College London, Royal Free Campus, London, UK
| | - Peter Kellman
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Theresa Munyombwe
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Chris P Gale
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds Teaching Hosptislas NHS Trust, Leeds, UK
| | - David L Buckley
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - John P Greenwood
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter P Swoboda
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
2
|
Wang T, Zhou D, Hong Z. Adipose tissue in older individuals: a contributing factor to sarcopenia. Metabolism 2024; 160:155998. [PMID: 39128607 DOI: 10.1016/j.metabol.2024.155998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Sarcopenia is a geriatric syndrome characterized by a functional decline in muscle. The prevalence of sarcopenia increases with natural aging, becoming a serious health problem among elderly individuals. Therefore, understanding the pathology of sarcopenia is critical for inhibiting age-related alterations and promoting health and longevity in elderly individuals. The development of sarcopenia may be influenced by interactions between visceral and subcutaneous adipose tissue and skeletal muscle, particularly under conditions of chronic low-grade inflammation and metabolic dysfunction. This hypothesis is supported by the following observations: (i) accumulation of senescent cells in both adipose tissue and skeletal muscle with age; (ii) gut dysbiosis, characterized by an imbalance in gut microbial communities as the main trigger for inflammation, sarcopenia, and aged adipose tissue; and (iii) microbial dysbiosis, which could impact the onset or progression of a senescent state. Moreover, adipose tissue acts as an endocrine organ, releasing molecules that participate in intricate communication networks between organs. Our discussion focuses on novel adipokines and their role in regulating adipose tissue and muscle, particularly those influenced by aging and obesity, emphasizing their contributions to disease development. On the basis of these findings, we propose that age-related adipose tissue and sarcopenia are disorders characterized by chronic inflammation and metabolic dysregulation. Finally, we explore new potential therapeutic strategies involving specialized proresolving mediator (SPM) G protein-coupled receptor (GPCR) agonists, non-SPM GPCR agonists, transient receptor potential (TRP) channels, antidiabetic drugs in conjunction with probiotics and prebiotics, and compounds designed to target senescent cells and mitigate their pro-inflammatory activity.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| | - Dong Zhou
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Mohamed S. Metformin: Diverse molecular mechanisms, gastrointestinal effects and overcoming intolerance in type 2 Diabetes Mellitus: A review. Medicine (Baltimore) 2024; 103:e40221. [PMID: 39470509 PMCID: PMC11521032 DOI: 10.1097/md.0000000000040221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Metformin, the first line treatment for patients with type 2 diabetes mellitus, has alternative novel roles, including cancer and diabetes prevention. This narrative review aims to explore its diverse mechanisms, effects and intolerance, using sources obtained by searching Scopus, PubMed and Web of Science databases, and following Scale for the Assessment of Narrative Review Articles reporting guidelines. Metformin exerts it actions through duration influenced, and organ specific, diverse mechanisms. Its use is associated with inhibition of hepatic gluconeogenesis targeted by mitochondria and lysosomes, reduction of cholesterol levels involving brown adipose tissue, weight reduction influenced by growth differentiation factor 15 and novel commensal bacteria, in addition to counteraction of meta-inflammation alongside immuno-modulation. Interactions with the gastrointestinal tract include alteration of gut microbiota, enhancement of glucose uptake and glucagon like peptide 1 and reduction of bile acid absorption. Though beneficial, they may be linked to intolerance. Metformin related gastrointestinal adverse effects are associated with dose escalation, immediate release formulations, gut microbiota alteration, epigenetic predisposition, inhibition of organic cation transporters in addition to interactions with serotonin, histamine and the enterohepatic circulation. Potentially effective measures to overcome intolerance encompasses carefully objective targeted dose escalation, prescription of fixed dose combination, microbiome modulators and prebiotics, in addition to use of extended release formulations.
Collapse
Affiliation(s)
- Sami Mohamed
- Department of Clinical Sciences, Dubai Medical University, Dubai, United Arab Emirates
| |
Collapse
|
4
|
Zhang B, Silverman S, Schwammn LH, Ji X, Singhal AB. Effectiveness of metformin pretreatment for stroke severity: A propensity score matching study. CNS Neurosci Ther 2024; 30:e70004. [PMID: 39169599 PMCID: PMC11339120 DOI: 10.1111/cns.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Metformin pretreatment might have neuroprotective effects. We aimed to determine the therapeutic effects of the antidiabetic medication metformin on ischemic stroke severity and discharge outcomes. METHODS We analyzed data on 1303 ischemic stroke patients who were on antidiabetic medications from the Massachusetts General Hospital (MGH) Advanced Comprehensive Stroke Center dataset (n = 8943, 2012-2022). We applied propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) analyses to investigate the effect of current usage of metformin (versus alternate antidiabetic treatment) on acute stroke clinical severity and discharge outcomes. RESULTS Of the 1303 patients who were on antidiabetic medications at the time of stroke admission, 730 (56%) were taking metformin. Metformin users were younger and more frequently had hypertension, whereas less frequently had prior CAD, AFib, and chronic kidney disease. The clinical features and laboratory values of the two groups were evenly distributed after PSM. Metformin-treated patients had statistically significant lower stroke severity on admission [National Institutes of Health Stroke Scale (NIHSS) (median, interquartile range) 3.0 (1.0-8.0) vs. 4.0 (2.0-11.3), p = 0.011], better functional independence at discharge (modified Rankin scale score 0-2, 36.3% vs. 25.4%, p < 0.001) and less in-hospital mortality (4.5% vs. 11.3%, p = 0.018). IPTW analysis results were consistent with PSM results. CONCLUSIONS Among diabetic patients with acute ischemic stroke, metformin appears to confer neuroprotection. Our results extend previous findings to the general stroke population. Stroke patients with diabetes mellitus who were treated with metformin prior to stroke, even when combined with additional antidiabetic medications, experienced less severe strokes upon admission and had better functional outcomes during hospitalization.
Collapse
Affiliation(s)
- Bowei Zhang
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Scott Silverman
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Lee H. Schwammn
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of NeurologyYale School of MedicineNew HavenConnecticutUSA
| | - Xunming Ji
- Department of NeurosurgeryXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Aneesh B. Singhal
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
5
|
Chaudhary S, Kulkarni A. Metformin: Past, Present, and Future. Curr Diab Rep 2024; 24:119-130. [PMID: 38568468 DOI: 10.1007/s11892-024-01539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW This review provides the most recent update of metformin, a biguanide oral antihyperglycemic drug used as a first-line treatment in type 2 diabetes mellitus. RECENT FINDINGS Metformin continues to dominate in the world of antidiabetics, and its use will continue to rise because of its high efficiency and easy availability. Apart from type 2 diabetes, research is exploring its potential in other conditions such as cancer, memory loss, bone disorders, immunological diseases, and aging. Metformin is the most prescribed oral antidiabetic worldwide. It has been in practical use for the last six decades and continues to be the preferred drug for newly diagnosed type 2 diabetes mellitus. It reduces glucose levels by decreasing hepatic glucose production, reducing intestinal glucose absorption, and increasing insulin sensitivity. It can be used as monotherapy or combined with other antidiabetics like sulfonylureas, DPP-4 inhibitors, SGLT-2 inhibitors, or insulin, improving its efficacy. Metformin can be used once or twice daily, depending on requirements. Prolonged usage of metformin may lead to abdominal discomfort, deficiency of Vitamin B12, or lactic acidosis. It should be used carefully in patients with renal impairment. Recent studies have explored additional benefits of metformin in polycystic ovarian disease, gestational diabetes mellitus, cognitive disorders, and immunological diseases. However, more extensive studies are needed to confirm these additional benefits.
Collapse
|
6
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
7
|
Goglia U, Hasballa I, Teti C, Boschetti M, Ferone D, Albertelli M. Ianus Bifrons: The Two Faces of Metformin. Cancers (Basel) 2024; 16:1287. [PMID: 38610965 PMCID: PMC11011026 DOI: 10.3390/cancers16071287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The ancient Roman god Ianus was a mysterious divinity with two opposite faces, one looking at the past and the other looking to the future. Likewise, metformin is an "old" drug, with one side looking at the metabolic role and the other looking at the anti-proliferative mechanism; therefore, it represents a typical and ideal bridge between diabetes and cancer. Metformin (1,1-dimethylbiguanidine hydrochloride) is a drug that has long been in use for the treatment of type 2 diabetes mellitus, but recently evidence is growing about its potential use in other metabolic conditions and in proliferative-associated diseases. The aim of this paper is to retrace, from a historical perspective, the knowledge of this molecule, shedding light on the subcellular mechanisms of action involved in metabolism as well as cellular and tissue growth. The intra-tumoral pharmacodynamic effects of metformin and its possible role in the management of different neoplasms are evaluated and debated. The etymology of the name Ianus is probably from the Latin term ianua, which means door. How many new doors will this old drug be able to open?
Collapse
Affiliation(s)
- Umberto Goglia
- Endocrinology and Diabetology Unit, Local Health Authority CN1, 12100 Cuneo, Italy
| | - Iderina Hasballa
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (M.B.); (D.F.); (M.A.)
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genoa, Italy
| | - Claudia Teti
- Endocrinology and Diabetology Unit, Local Health Autorithy Imperia 1, 18100 Imperia, Italy;
| | - Mara Boschetti
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (M.B.); (D.F.); (M.A.)
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genoa, Italy
| | - Diego Ferone
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (M.B.); (D.F.); (M.A.)
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genoa, Italy
| | - Manuela Albertelli
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy (M.B.); (D.F.); (M.A.)
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genoa, Italy
| |
Collapse
|
8
|
Sawamoto A, Okada M, Matsuoka N, Okuyama S, Nakajima M. Tipepidine activates AMPK and improves adipose tissue fibrosis and glucose intolerance in high-fat diet-induced obese mice. FASEB J 2024; 38:e23542. [PMID: 38466234 DOI: 10.1096/fj.202301861rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
Tipepidine (3-[di-2-thienylmethylene]-1-methylpiperidine) (TP) is a non-narcotic antitussive used in Japan. Recently, the potential application of TP in the treatment of neuropsychiatric disorders, such as depression and attention deficit hyperactivity disorder, has been suggested; however, its functions in energy metabolism are unknown. Here, we demonstrate that TP exhibits a metabolism-improving action. The administration of TP reduced high-fat diet-induced body weight gain in mice and lipid accumulation in the liver and increased the weight of epididymal white adipose tissue (eWAT) in diet-induced obese (DIO) mice. Furthermore, TP inhibited obesity-induced fibrosis in the eWAT. We also found that TP induced AMP-activated protein kinase (AMPK) activation in the eWAT of DIO mice and 3T3-L1 cells. TP-induced AMPK activation was abrogated by the transfection of liver kinase B1 siRNA in 3T3-L1 cells. The metabolic effects of TP were almost equivalent to those of metformin, an AMPK activator that is used as a first-line antidiabetic drug. In summary, TP is a potent AMPK activator, suggesting its novel role as an antidiabetic drug owing to its antifibrotic effect on adipose tissues.
Collapse
Affiliation(s)
- Atsushi Sawamoto
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Madoka Okada
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Nanako Matsuoka
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Satoshi Okuyama
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| | - Mitsunari Nakajima
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Ehime, Japan
| |
Collapse
|
9
|
Alqarni M, Alshehri AA, Arida H. Validation and Application of Screen-Printed Microchip for Potentiometric Determination of Metformin Hydrochloride in Tablet Dosage Form. Int J Anal Chem 2024; 2024:8664723. [PMID: 38445178 PMCID: PMC10914426 DOI: 10.1155/2024/8664723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/29/2023] [Accepted: 02/18/2024] [Indexed: 03/07/2024] Open
Abstract
Metformin is an oral biguanides hypoglycaemic agent, which used to lower the blood glucose levels in people with type 2 diabetes mellitus. Many analytical techniques have been used to quantify the drug in different pharmaceutical dosage forms; however, most of these methods have limited throughput in the quality control application. A disposable potentiometric microsensor responsive to metformin has recently been reported. For the first time, herein, this method of analysis has been validated according to IUPAC recommendations and successfully applied in the determination of metformin drug in some dosage form. Different drug formulations of metformin hydrochloride have been collected from the local pharmaceutical stores in Saudi Arabia and analysed using the validated microchip-based method of analysis. Subsequently, the results of this study showed that the validated method was linear, specific, precise, and accurate. The linear range was 1 × 10-1-1 × 10-5 mol L-1 and the correlation coefficient was 0.999. The limit of detection was 2.89 × 10-6 mol L-1, and the limit of quantification was 8.77 × 10-6 mol L-1. This method demonstrated high precision, with an RSD% of less than 2.22%. The accuracy of this method was obtained by comparing the recovery percentage with percentage values less than 5%. The results obtained showed that there was no significant difference between the references, label, and recovery of less than 5%.
Collapse
Affiliation(s)
- Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abdullah A. Alshehri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hassan Arida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
10
|
Liang G, Fang J, Zhang P, Ding S, Zhao Y, Feng Y. Metformin plus L-carnitine enhances brown/beige adipose tissue activity via Nrf2/HO-1 signaling to reduce lipid accumulation and inflammation in murine obesity. Open Med (Wars) 2024; 19:20240900. [PMID: 38463531 PMCID: PMC10921440 DOI: 10.1515/med-2024-0900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/13/2023] [Accepted: 12/18/2023] [Indexed: 03/12/2024] Open
Abstract
This study investigated how Metformin (Met) combined with L-carnitine (L-car) modulates brown adipose tissue (BAT) to affect obesity. High-fat-induced obese rats received daily oral gavage with Met and/or L-car, followed by serum biochemical analysis, histopathological observation on adipose tissues, and immunochemistry test for the abdominal expression of BAT-specific uncoupling protein 1 (UCP1). Mouse-embryonic-fibroblast cells were induced into adipocytes, during which Met plus L-car was added with/without saturated fatty acid (SFA). The role of nuclear factor erythroid 2-related factor 2 (Nrf2) in adipocyte browning was investigated by gene silencing. Mitochondria biogenesis in adipocytes was inspected by Mitotracker staining. Nrf2/heme oxygenase-1 (HO-1)/BAT-related genes/proinflammatory marker expressions in adipose tissues and/or adipocytes were analyzed by Western blot, qRT-PCR, and/or immunofluorescence test. Met or L-car improved metabolic disorders, reduced adipocyte vacuolization and swelling, upregulated levels of BAT-related genes including UCP1 and downregulated proinflammatory marker expressions, and activated the Nrf2/HO-1 pathway in adipose tissues of obese rats. Met and L-car functioned more strongly than alone. In adipocytes, Met plus L-car upregulated BAT-related gene levels and protected against SFA-caused inflammation promotion and mitochondria degeneration, which yet was attenuated by Nrf2 silencing. Met plus L-car enhances BAT activity and white adipose tissue browning via the Nrf2/HO-1 pathway to reduce lipid accumulation and inflammation in obese rats.
Collapse
Affiliation(s)
- Guojin Liang
- Anesthesiology Department, Ningbo First Hospital, Ningbo, China
| | - Jie Fang
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Pingping Zhang
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Shuxia Ding
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Yudan Zhao
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Yueying Feng
- Paediatrics Department, Ningbo Women and Children’s Hospital, No. 339 Liuting Street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
11
|
Foretz M, Guigas B, Viollet B. Metformin: update on mechanisms of action and repurposing potential. Nat Rev Endocrinol 2023; 19:460-476. [PMID: 37130947 PMCID: PMC10153049 DOI: 10.1038/s41574-023-00833-4] [Citation(s) in RCA: 178] [Impact Index Per Article: 178.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/04/2023]
Abstract
Currently, metformin is the first-line medication to treat type 2 diabetes mellitus (T2DM) in most guidelines and is used daily by >200 million patients. Surprisingly, the mechanisms underlying its therapeutic action are complex and are still not fully understood. Early evidence highlighted the liver as the major organ involved in the effect of metformin on reducing blood levels of glucose. However, increasing evidence points towards other sites of action that might also have an important role, including the gastrointestinal tract, the gut microbial communities and the tissue-resident immune cells. At the molecular level, it seems that the mechanisms of action vary depending on the dose of metformin used and duration of treatment. Initial studies have shown that metformin targets hepatic mitochondria; however, the identification of a novel target at low concentrations of metformin at the lysosome surface might reveal a new mechanism of action. Based on the efficacy and safety records in T2DM, attention has been given to the repurposing of metformin as part of adjunct therapy for the treatment of cancer, age-related diseases, inflammatory diseases and COVID-19. In this Review, we highlight the latest advances in our understanding of the mechanisms of action of metformin and discuss potential emerging novel therapeutic uses.
Collapse
Affiliation(s)
- Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France.
| |
Collapse
|
12
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
13
|
Ziqubu K, Mazibuko-Mbeje SE, Mthembu SXH, Mabhida SE, Jack BU, Nyambuya TM, Nkambule BB, Basson AK, Tiano L, Dludla PV. Anti-Obesity Effects of Metformin: A Scoping Review Evaluating the Feasibility of Brown Adipose Tissue as a Therapeutic Target. Int J Mol Sci 2023; 24:ijms24032227. [PMID: 36768561 PMCID: PMC9917329 DOI: 10.3390/ijms24032227] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Brown adipose tissue (BAT) is increasingly recognized as the major therapeutic target to promote energy expenditure and ameliorate diverse metabolic complications. There is a general interest in understanding the pleiotropic effects of metformin against metabolic complications. Major electronic databases and search engines such as PubMed/MEDLINE, Google Scholar, and the Cochrane library were used to retrieve and critically discuss evidence reporting on the impact of metformin on regulating BAT thermogenic activity to ameliorate complications linked with obesity. The summarized evidence suggests that metformin can reduce body weight, enhance insulin sensitivity, and improve glucose metabolism by promoting BAT thermogenic activity in preclinical models of obesity. Notably, this anti-diabetic agent can affect the expression of major thermogenic transcriptional factors such as uncoupling protein 1 (UCP1), nuclear respiratory factor 1 (NRF1), and peroxisome-proliferator-activated receptor gamma coactivator 1-alpha (PGC1-α) to improve BAT mitochondrial function and promote energy expenditure. Interestingly, vital molecular markers involved in glucose metabolism and energy regulation such as AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21) are similarly upregulated by metformin treatment in preclinical models of obesity. The current review also discusses the clinical relevance of BAT and thermogenesis as therapeutic targets. This review explored critical components including effective dosage and appropriate intervention period, consistent with the beneficial effects of metformin against obesity-associated complications.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Sithandiwe E. Mazibuko-Mbeje
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
- Correspondence: (S.E.M.-M.); (P.V.D.); Tel.: +27-21-938-0333 (P.V.D.)
| | - Sinenhlanhla X. H. Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Sihle E. Mabhida
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Babalwa U. Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Tawanda M. Nyambuya
- Department of Health Sciences, Namibia University of Science and Technology, Windhoek 9000, Namibia
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Albertus K. Basson
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Phiwayinkosi V. Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
- Correspondence: (S.E.M.-M.); (P.V.D.); Tel.: +27-21-938-0333 (P.V.D.)
| |
Collapse
|
14
|
“Ferrocrinology”—Iron Is an Important Factor Involved in Gluco- and Lipocrinology. Nutrients 2022; 14:nu14214693. [DOI: 10.3390/nu14214693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
“Ferrocrinology” is the term used to describe the study of iron effects on the functioning of adipose tissue, which together with muscle tissue makes the largest endocrine organ in the human body. By impairing exercise capacity, reducing AMP-activated kinase activity, and enhancing insulin resistance, iron deficiency can lead to the development of obesity and type 2 diabetes mellitus. Due to impaired browning of white adipose tissue and reduced mitochondrial iron content in adipocytes, iron deficiency (ID) can cause dysfunction of brown adipose tissue. By reducing ketogenesis, aconitase activity, and total mitochondrial capacity, ID impairs muscle performance. Another important aspect is the effect of ID on the impairment of thermogenesis due to reduced binding of thyroid hormones to their nuclear receptors, with subsequently impaired utilization of norepinephrine in tissues, and impaired synthesis and distribution of cortisol, which all make the body’s reactivity to stress in ID more pronounced. Iron deficiency can lead to the development of the most common endocrinopathy, autoimmune thyroid disease. In this paper, we have discussed the role of iron in the cross-talk between glucocrinology, lipocrinology and myocrinology, with thyroid hormones acting as an active bystander.
Collapse
|
15
|
Markussen LK, Rondini EA, Johansen OS, Madsen JGS, Sustarsic EG, Marcher AB, Hansen JB, Gerhart-Hines Z, Granneman JG, Mandrup S. Lipolysis regulates major transcriptional programs in brown adipocytes. Nat Commun 2022; 13:3956. [PMID: 35803907 PMCID: PMC9270495 DOI: 10.1038/s41467-022-31525-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
β-Adrenergic signaling is a core regulator of brown adipocyte function stimulating both lipolysis and transcription of thermogenic genes, thereby expanding the capacity for oxidative metabolism. We have used pharmacological inhibitors and a direct activator of lipolysis to acutely modulate the activity of lipases, thereby enabling us to uncover lipolysis-dependent signaling pathways downstream of β-adrenergic signaling in cultured brown adipocytes. Here we show that induction of lipolysis leads to acute induction of several gene programs and is required for transcriptional regulation by β-adrenergic signals. Using machine-learning algorithms to infer causal transcription factors, we show that PPARs are key mediators of lipolysis-induced activation of genes involved in lipid metabolism and thermogenesis. Importantly, however, lipolysis also activates the unfolded protein response and regulates the core circadian transcriptional machinery independently of PPARs. Our results demonstrate that lipolysis generates important metabolic signals that exert profound pleiotropic effects on transcription and function of cultured brown adipocytes.
Collapse
Affiliation(s)
- Lasse K Markussen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Center for Adipocyte Signaling (AdipoSign), Odense, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Odense, Denmark
| | - Elizabeth A Rondini
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Olivia Sveidahl Johansen
- Center for Adipocyte Signaling (AdipoSign), Odense, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Biotech ApS, Copenhagen, Denmark
| | - Jesper G S Madsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Odense, Denmark
| | - Elahu G Sustarsic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Britt Marcher
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
- Center for Adipocyte Signaling (AdipoSign), Odense, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Odense, Denmark
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Zachary Gerhart-Hines
- Center for Adipocyte Signaling (AdipoSign), Odense, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Embark Biotech ApS, Copenhagen, Denmark
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
- Center for Adipocyte Signaling (AdipoSign), Odense, Denmark.
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Odense, Denmark.
| |
Collapse
|
16
|
Transdermal Delivery of Metformin Using Dissolving Microneedles and Iontophoresis Patches for Browning Subcutaneous Adipose Tissue. Pharmaceutics 2022; 14:pharmaceutics14040879. [PMID: 35456713 PMCID: PMC9029293 DOI: 10.3390/pharmaceutics14040879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is a serious public health problem that is strongly associated with increased multiple comorbidities such as diabetes, cardiovascular disease, and some types of cancer. While current anti-obesity treatments have various issues, locally transforming energy-storing white adipose tissue (WAT) into energy-burning brown-like/beige adipose tissue, the so-called browning of WAT, has been suggested to enhance obesity treatment efficiency with minimized side effects. Metformin is a first-line antidiabetes drug and a potent activator of AMP-activated protein kinase. Emerging evidence has suggested that metformin might enhance energy expenditure via the browning of WAT and hence reduce body weight. Subcutaneous WAT is easier to access and has a stronger browning potential than other WAT depots. In this study, we used dissolvable poly (lactic-co-glycolic acid) microneedles (MN) to deliver metformin to the subcutaneous WAT in obese C57BL/6J mice with the assistance of iontophoresis (INT), and then investigated metformin-induced WAT browning and its subsequent thermogenesis effects. Compared with MN alone or INT alone, MN + INT had better anti-obesity activity, as indicated by decreasing body weight and fat gain, increased energy expenditure, decreased fat pad size, and improved energy metabolism through the browning of WAT. Browning subcutaneous WAT by delivering metformin and other browning agents using this MN + INT approach might combat obesity in an effective, easy, and safe regimen.
Collapse
|
17
|
Rosina M, Ceci V, Turchi R, Chuan L, Borcherding N, Sciarretta F, Sánchez-Díaz M, Tortolici F, Karlinsey K, Chiurchiù V, Fuoco C, Giwa R, Field RL, Audano M, Arena S, Palma A, Riccio F, Shamsi F, Renzone G, Verri M, Crescenzi A, Rizza S, Faienza F, Filomeni G, Kooijman S, Rufini S, de Vries AAF, Scaloni A, Mitro N, Tseng YH, Hidalgo A, Zhou B, Brestoff JR, Aquilano K, Lettieri-Barbato D. Ejection of damaged mitochondria and their removal by macrophages ensure efficient thermogenesis in brown adipose tissue. Cell Metab 2022; 34:533-548.e12. [PMID: 35305295 PMCID: PMC9039922 DOI: 10.1016/j.cmet.2022.02.016] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/11/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023]
Abstract
Recent findings have demonstrated that mitochondria can be transferred between cells to control metabolic homeostasis. Although the mitochondria of brown adipocytes comprise a large component of the cell volume and undergo reorganization to sustain thermogenesis, it remains unclear whether an intercellular mitochondrial transfer occurs in brown adipose tissue (BAT) and regulates adaptive thermogenesis. Herein, we demonstrated that thermogenically stressed brown adipocytes release extracellular vesicles (EVs) that contain oxidatively damaged mitochondrial parts to avoid failure of the thermogenic program. When re-uptaken by parental brown adipocytes, mitochondria-derived EVs reduced peroxisome proliferator-activated receptor-γ signaling and the levels of mitochondrial proteins, including UCP1. Their removal via the phagocytic activity of BAT-resident macrophages is instrumental in preserving BAT physiology. Depletion of macrophages in vivo causes the abnormal accumulation of extracellular mitochondrial vesicles in BAT, impairing the thermogenic response to cold exposure. These findings reveal a homeostatic role of tissue-resident macrophages in the mitochondrial quality control of BAT.
Collapse
Affiliation(s)
- Marco Rosina
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Neurology Unit, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Veronica Ceci
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Riccardo Turchi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Li Chuan
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | | - María Sánchez-Díaz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Flavia Tortolici
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Keaton Karlinsey
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Valerio Chiurchiù
- IRCCS, Fondazione Santa Lucia, 00179 Rome, Italy; Institute of Translational Pharmacology, Laboratory of Resolution of Neuroinflammation, National Research Council, 00133 Rome, Italy
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Rocky Giwa
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Rachael L Field
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Simona Arena
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM-National Research Council, Portici, 80055 Naples, Italy
| | - Alessandro Palma
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Giovanni Renzone
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM-National Research Council, Portici, 80055 Naples, Italy
| | - Martina Verri
- Pathology Unit, University Hospital Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Anna Crescenzi
- Pathology Unit, University Hospital Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Salvatore Rizza
- Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
| | | | | | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Stefano Rufini
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antoine A F de Vries
- Department of Cardiology, Laboratory of Experimental Cardiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM-National Research Council, Portici, 80055 Naples, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06030, USA
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; IRCCS, Fondazione Santa Lucia, 00179 Rome, Italy.
| |
Collapse
|
18
|
Intestinal AMPK modulation of microbiota mediates crosstalk with brown fat to control thermogenesis. Nat Commun 2022; 13:1135. [PMID: 35241650 PMCID: PMC8894485 DOI: 10.1038/s41467-022-28743-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/02/2022] [Indexed: 01/28/2023] Open
Abstract
The energy-dissipating capacity of brown adipose tissue through thermogenesis can be targeted to improve energy balance. Mammalian 5'-AMP-activated protein kinase, a key nutrient sensor for maintaining cellular energy status, is a known therapeutic target in Type II diabetes. Despite its well-established roles in regulating glucose metabolism in various tissues, the functions of AMPK in the intestine remain largely unexplored. Here we show that AMPKα1 deficiency in the intestine results in weight gain and impaired glucose tolerance under high fat diet feeding, while metformin administration fails to ameliorate these metabolic disorders in intestinal AMPKα1 knockout mice. Further, AMPKα1 in the intestine communicates with brown adipose tissue to promote thermogenesis. Mechanistically, we uncover a link between intestinal AMPKα1 activation and BAT thermogenic regulation through modulating anti-microbial peptide-controlled gut microbiota and the metabolites. Our findings identify AMPKα1-mediated mechanisms of intestine-BAT communication that may partially underlie the therapeutic effects of metformin.
Collapse
|
19
|
Huang Y, Peng T, Hu W, Gao X, Chen Y, Zhang Q, Wu C, Pan X. Fully armed photodynamic therapy with spear and shear for topical deep hypertrophic scar treatment. J Control Release 2022; 343:408-419. [DOI: 10.1016/j.jconrel.2022.01.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 12/25/2022]
|
20
|
Berberine and lycopene as alternative or add-on therapy to metformin and statins, a review. Eur J Pharmacol 2021; 913:174590. [PMID: 34801530 DOI: 10.1016/j.ejphar.2021.174590] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022]
Abstract
Nutraceuticals are principally extracted from natural products that are frequently safe and well-tolerated. Lycopene and berberine are natural plants with a wide range of beneficial effects including protective activities against metabolic disorders such as diabetes and cardiovascular diseases. These compounds might be considered technically more as a drug than a nutraceutical and could be prescribed as a product. However, further studies are needed to understand if these supplements could affect metabolic syndrome outcomes. Even if nutraceuticals exert a prophylactic activity within the body, their bioactivity and bioavailability have high interindividual variation, and precise assessment of biological function of these bioactive compounds in randomized clinical trials is critical. However, these reports must be interpreted with more considerations due to the low quality of the trials. The aim of this paper is to bring evidence about the management of cardiovascular diseases and diabetes through the use of nutraceuticals with particular attention to lycopene and berberine effectiveness.
Collapse
|
21
|
Stojnić B, Serrano A, Sušak L, Palou A, Bonet ML, Ribot J. Protective Effects of Individual and Combined Low Dose Beta-Carotene and Metformin Treatments against High-Fat Diet-Induced Responses in Mice. Nutrients 2021; 13:3607. [PMID: 34684608 PMCID: PMC8538788 DOI: 10.3390/nu13103607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022] Open
Abstract
Anti-obesity activity has been reported for beta-carotene (BC) supplementation at high doses and metformin (MET). We studied whether BC treatment at a closer to dietary dose and MET treatment at a lower than therapeutic dose are effective in ameliorating unwanted effects of an obesogenic diet and whether their combination is advantageous. Obesity-prone mice were challenged with a high-fat diet (HFD, 45% energy as fat) for 4 weeks while receiving a placebo or being treated orally with BC (3 mg/kg/day), MET (100 mg/kg/day), or their combination (BC+MET); a fifth group received a placebo and was kept on a normal-fat diet (10% energy as fat). HFD-induced increases in body weight gain and inguinal white adipose tissue (WAT) adipocyte size were attenuated maximally or selectively in the BC+MET group, in which a redistribution towards smaller adipocytes was noted. Cumulative energy intake was unaffected, yet results suggested increased systemic energy expenditure and brown adipose tissue activation in the treated groups. Unwanted effects of HFD on glucose control and insulin sensitivity were attenuated in the treated groups, especially BC and BC+MET, in which hepatic lipid content was also decreased. Transcriptional analyses suggested effects on skeletal muscle and WAT metabolism could contribute to better responses to the HFD, especially in the MET and BC+MET groups. The results support the benefits of the BC+MET cotreatment.
Collapse
Affiliation(s)
- Bojan Stojnić
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
| | - Alba Serrano
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
| | - Lana Sušak
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
| | - Andreu Palou
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 07122 Palma, Spain
| | - M. Luisa Bonet
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 07122 Palma, Spain
| | - Joan Ribot
- Grupo de Nutrigenómica, Biomarcadores y Evaluación de Riesgos, Laboratory of Molecular Biology, Nutrition and Biotechnology (LBNB), Universitat de les Illes Balears, 07122 Palma, Spain; (B.S.); (A.S.); (L.S.); (A.P.); (J.R.)
- Institut d’Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 07122 Palma, Spain
| |
Collapse
|
22
|
Metformin activates chaperone-mediated autophagy and improves disease pathologies in an Alzheimer disease mouse model. Protein Cell 2021; 12:769-787. [PMID: 34291435 PMCID: PMC8464644 DOI: 10.1007/s13238-021-00858-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/03/2021] [Indexed: 01/15/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is a lysosome-dependent selective degradation pathway implicated in the pathogenesis of cancer and neurodegenerative diseases. However, the mechanisms that regulate CMA are not fully understood. Here, using unbiased drug screening approaches, we discover Metformin, a drug that is commonly the first medication prescribed for type 2 diabetes, can induce CMA. We delineate the mechanism of CMA induction by Metformin to be via activation of TAK1-IKKα/β signaling that leads to phosphorylation of Ser85 of the key mediator of CMA, Hsc70, and its activation. Notably, we find that amyloid-beta precursor protein (APP) is a CMA substrate and that it binds to Hsc70 in an IKKα/β-dependent manner. The inhibition of CMA-mediated degradation of APP enhances its cytotoxicity. Importantly, we find that in the APP/PS1 mouse model of Alzheimer’s disease (AD), activation of CMA by Hsc70 overexpression or Metformin potently reduces the accumulated brain Aβ plaque levels and reverses the molecular and behavioral AD phenotypes. Our study elucidates a novel mechanism of CMA regulation via Metformin-TAK1-IKKα/β-Hsc70 signaling and suggests Metformin as a new activator of CMA for diseases, such as AD, where such therapeutic intervention could be beneficial.
Collapse
|
23
|
Hoffman JM, Valencak TG. Sex differences and aging: Is there a role of brown adipose tissue? Mol Cell Endocrinol 2021; 531:111310. [PMID: 33989715 PMCID: PMC8195864 DOI: 10.1016/j.mce.2021.111310] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/31/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022]
Abstract
In every population across the world, women live significantly longer than men; however, the underlying physiological processes that drive these sex differences in age-specific mortality are largely unknown. Recently, the role of adipose tissue in aging and longevity has been a focus of biomedical research in both humans and rodent models. Specifically, brown adipose tissue, a thermoregulatory tissue originally thought to not exist past infancy in humans, has been shown to potentially play a role in health throughout the lifespan. Females have larger adult brown adipose depots that are not just larger in size but also more efficient in non-shivering thermogenesis. This improved functioning of the brown adipose tissue may potentially lead to improved female health, and we hypothesize that this advantage may be of even bigger significance in the older population. Here, we briefly review what is known about sex differences in aging and how sex differences in brown adipose tissue may be contributing to the female lifespan advantage. These questions have usually been addressed in large experimental studies in rodents as a translational model of human aging. Overall, we propose that a better understanding of the thermogenesis-metabolism nexus is necessary in biomedical research, and sex differences in these factors may contribute to the female longevity bias seen in human populations.
Collapse
Affiliation(s)
- Jessica M Hoffman
- Department of Biology, University of Alabama at Birmingham, 1300 University Blvd., CH464, Birmingham, AL, 35294, USA.
| | - Teresa G Valencak
- College of Animal Sciences, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, 310058, Hangzhou, PR China.
| |
Collapse
|
24
|
The Hormetic Effect of Metformin: "Less Is More"? Int J Mol Sci 2021; 22:ijms22126297. [PMID: 34208371 PMCID: PMC8231127 DOI: 10.3390/ijms22126297] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Metformin (MTF) is the first-line therapy for type 2 diabetes (T2DM). The euglycemic effect of MTF is due to the inhibition of hepatic glucose production. Literature reports that the principal molecular mechanism of MTF is the activation of 5′-AMP-activated protein kinase (AMPK) due to the decrement of ATP intracellular content consequent to the inhibition of Complex I, although this effect is obtained only at millimolar concentrations. Conversely, micromolar MTF seems to activate the mitochondrial electron transport chain, increasing ATP production and limiting oxidative stress. This evidence sustains the idea that MTF exerts a hormetic effect based on its concentration in the target tissue. Therefore, in this review we describe the effects of MTF on T2DM on the principal target organs, such as liver, gut, adipose tissue, endothelium, heart, and skeletal muscle. In particular, data indicate that all organs, except the gut, accumulate MTF in the micromolar range when administered in therapeutic doses, unmasking molecular mechanisms that do not depend on Complex I inhibition.
Collapse
|
25
|
Morais T, Seabra AL, Patrício BG, Guimarães M, Nora M, Oliveira PF, Alves MG, Monteiro MP. Visceral Adipose Tissue Displays Unique Metabolomic Fingerprints in Obesity, Pre-Diabetes and Type 2 Diabetes. Int J Mol Sci 2021; 22:5695. [PMID: 34071774 PMCID: PMC8199212 DOI: 10.3390/ijms22115695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022] Open
Abstract
Visceral adipose tissue (VAT) metabolic profiling harbors the potential to disentangle molecular changes underlying obesity-related dysglycemia. In this study, the VAT exometabolome of subjects with obesity and different glycemic statuses are analyzed. The subjects (n = 19) are divided into groups according to body mass index and glycemic status: subjects with obesity and euglycemia (Ob+NGT, n = 5), subjects with obesity and pre-diabetes (Ob+Pre-T2D, n = 5), subjects with obesity and type 2 diabetes under metformin treatment (Ob+T2D, n = 5) and subjects without obesity and with euglycemia (Non-Ob, n = 4), used as controls. VATs are incubated in culture media and extracellular metabolite content is determined by proton nuclear magnetic resonance (1H-NMR). Glucose consumption is not different between the groups. Pyruvate and pyroglutamate consumption are significantly lower in all groups of subjects with obesity compared to Non-Ob, and significantly lower in Ob+Pre-T2D as compared to Ob+NGT. In contrast, isoleucine consumption is significantly higher in all groups of subjects with obesity, particularly in Ob+Pre-T2D, compared to Non-Ob. Acetate production is also significantly lower in Ob+Pre-T2D compared to Non-Ob. In sum, the VAT metabolic fingerprint is associated with pre-diabetes and characterized by higher isoleucine consumption, accompanied by lower acetate production and pyruvate and pyroglutamate consumption. We propose that glucose metabolism follows different fates within the VAT, depending on the individuals' health status.
Collapse
Affiliation(s)
- Tiago Morais
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Alexandre L. Seabra
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Bárbara G. Patrício
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Marta Guimarães
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Mário Nora
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Pedro F. Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Marco G. Alves
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Mariana P. Monteiro
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; (T.M.); (A.L.S.); (B.G.P.); (M.G.); (M.N.); (M.G.A.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
26
|
Hammoud SH, AlZaim I, Mougharbil N, Koubar S, Eid AH, Eid AA, El-Yazbi AF. Peri-renal adipose inflammation contributes to renal dysfunction in a non-obese prediabetic rat model: Role of anti-diabetic drugs. Biochem Pharmacol 2021; 186:114491. [PMID: 33647265 DOI: 10.1016/j.bcp.2021.114491] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/07/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy is a major health challenge with considerable economic burden and significant impact on patients' quality of life. Despite recent advances in diabetic patient care, current clinical practice guidelines fall short of halting the progression of diabetic nephropathy to end-stage renal disease. Moreover, prior literature reported manifestations of renal dysfunction in early stages of metabolic impairment prior to the development of hyperglycemia indicating the involvement of alternative pathological mechanisms apart from those typically triggered by high blood glucose. Here, we extend our prior research work implicating localized inflammation in specific adipose depots in initiating cardiovascular dysfunction in early stages of metabolic impairment. Non-obese prediabetic rats showed elevated glomerular filtration rates and mild proteinuria in absence of hyperglycemia, hypertension, and signs of systemic inflammation. Isolated perfused kidneys from these rats showed impaired renovascular endothelial feedback in response to vasopressors and increased flow. While endothelium dependent dilation remained functional, renovascular relaxation in prediabetic rats was not mediated by nitric oxide and prostaglandins as in control tissues, but rather an upregulation of the function of epoxy eicosatrienoic acids was observed. This was coupled with signs of peri-renal adipose tissue (PRAT) inflammation and renal structural damage. A two-week treatment with non-hypoglycemic doses of metformin or pioglitazone, shown previously to ameliorate adipose inflammation, not only reversed PRAT inflammation in prediabetic rats, but also reversed the observed functional, renovascular, and structural renal abnormalities. The present results suggest that peri-renal adipose inflammation triggers renal dysfunction early in the course of metabolic disease.
Collapse
Affiliation(s)
- Safaa H Hammoud
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Nahed Mougharbil
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Sahar Koubar
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Assaad A Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon.
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt.
| |
Collapse
|
27
|
Wang L, Liu D, Wei G, Ge H. Berberine and Metformin in the Treatment of Type 2 Diabetes Mellitus: A Systemic Review and Meta-Analysis of Randomized Clinical Trials. Health (London) 2021. [DOI: 10.4236/health.2021.1311096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Demaré S, Kothari A, Calcutt NA, Fernyhough P. Metformin as a potential therapeutic for neurological disease: mobilizing AMPK to repair the nervous system. Expert Rev Neurother 2020; 21:45-63. [PMID: 33161784 PMCID: PMC9482886 DOI: 10.1080/14737175.2021.1847645] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Metformin is currently first line therapy for type 2 diabetes (T2D). The mechanism of action of metformin involves activation of AMP-activated protein kinase (AMPK) to enhance mitochondrial function (for example, biogenesis, refurbishment and dynamics) and autophagy. Many neurodegenerative diseases of the central and peripheral nervous systems arise from metabolic failure and toxic protein aggregation where activated AMPK could prove protective. Areas covered: The authors review literature on metformin treatment in Parkinson’s disease, Huntington’s disease and other neurological diseases of the CNS along with neuroprotective effects of AMPK activation and suppression of the mammalian target of rapamycin (mTOR) pathway on peripheral neuropathy and neuropathic pain. The authors compare the efficacy of metformin with the actions of resveratrol. Expert opinion: Metformin, through activation of AMPK and autophagy, can enhance neuronal bioenergetics, promote nerve repair and reduce toxic protein aggregates in neurological diseases. A long history of safe use in humans should encourage development of metformin and other AMPK activators in preclinical and clinical research. Future studies in animal models of neurological disease should strive to further dissect in a mechanistic manner the pathways downstream from metformin-dependent AMPK activation, and to further investigate mTOR dependent and independent signaling pathways driving neuroprotection.
Collapse
Affiliation(s)
- Sarah Demaré
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| | - Asha Kothari
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego , La Jolla, CA, USA
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre , Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba , Winnipeg, MB, Canada
| |
Collapse
|
29
|
Oliveira FR, Mamede M, Bizzi MF, Rocha ALL, Ferreira CN, Gomes KB, Cândido AL, Reis FM. Effects of Short Term Metformin Treatment on Brown Adipose Tissue Activity and Plasma Irisin Levels in Women with Polycystic Ovary Syndrome: A Randomized Controlled Trial. Horm Metab Res 2020; 52:718-723. [PMID: 32365398 DOI: 10.1055/a-1157-0615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a chronic dysfunction associated with obesity and metabolic disorders that can be ameliorated by treatment with metformin. Brown adipose tissue (BAT) has been recently identified in adult humans, and irisin is a myokine that induces BAT formation. The aim of this randomized controlled trial was to evaluate whether a short term treatment with metformin alters BAT activity and plasma irisin levels in women with PCOS. The participants were randomly assigned to receive metformin (1500 mg/day, n=21) or placebo (n=24) during 60 days. BAT activity was assessed by 18F-FDG positron emission tomography-computed tomography (PET-CT) and plasma irisin levels were measured by enzyme immunoassay. The groups were similar in age, body measures, metabolic profile and PCOS phenotypes. BAT activity did not change significantly in the women treated with metformin (median Δ SUVmax=-0.06 g/ml, interquartile interval -2.81 to 0.24 g/ml, p=0.484, Wilcoxon's test) or placebo (median Δ SUVmax=0.98 g/ml, interquartile interval -2.94 to 4.60 g/ml, p=0.386). In addition, plasma irisin levels remained unchanged in the groups treated with metformin (median Δ=-98 ng/ml, interquartile interval -366 to 60 ng/ml, p=0.310) and placebo (median Δ=28 ng/ml, interquartile interval -1260 to 215 ng/ml, p=0.650). These results suggest that in PCOS women BAT activity and plasma irisin levels may not change after a brief treatment with metformin.
Collapse
Affiliation(s)
- Flávia R Oliveira
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Odete Valadares Maternity Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Mamede
- Department of Anatomy and Imaging, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana F Bizzi
- Department of Internal Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Luiza L Rocha
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cláudia N Ferreira
- Technical College, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karina B Gomes
- Clinical and Toxicological Analyses, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana L Cândido
- Department of Internal Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando M Reis
- Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
30
|
Abstract
The organic cation transporters (OCTs) OCT1, OCT2, OCT3, novel OCT (OCTN)1, OCTN2, multidrug and toxin exclusion (MATE)1, and MATE kidney-specific 2 are polyspecific transporters exhibiting broadly overlapping substrate selectivities. They transport organic cations, zwitterions, and some uncharged compounds and operate as facilitated diffusion systems and/or antiporters. OCTs are critically involved in intestinal absorption, hepatic uptake, and renal excretion of hydrophilic drugs. They modulate the distribution of endogenous compounds such as thiamine, L-carnitine, and neurotransmitters. Sites of expression and functions of OCTs have important impact on energy metabolism, pharmacokinetics, and toxicity of drugs, and on drug-drug interactions. In this work, an overview about the human OCTs is presented. Functional properties of human OCTs, including identified substrates and inhibitors of the individual transporters, are described. Sites of expression are compiled, and data on regulation of OCTs are presented. In addition, genetic variations of OCTs are listed, and data on their impact on transport, drug treatment, and diseases are reported. Moreover, recent data are summarized that indicate complex drug-drug interaction at OCTs, such as allosteric high-affinity inhibition of transport and substrate dependence of inhibitor efficacies. A hypothesis about the molecular mechanism of polyspecific substrate recognition by OCTs is presented that is based on functional studies and mutagenesis experiments in OCT1 and OCT2. This hypothesis provides a framework to imagine how observed complex drug-drug interactions at OCTs arise. Finally, preclinical in vitro tests that are performed by pharmaceutical companies to identify interaction of novel drugs with OCTs are discussed. Optimized experimental procedures are proposed that allow a gapless detection of inhibitory and transported drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 2020; 134:827-851. [PMID: 32271386 DOI: 10.1042/cs20190227] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Major shifts in human lifestyle and dietary habits toward sedentary behavior and refined food intake triggered steep increase in the incidence of metabolic disorders including obesity and Type 2 diabetes. Patients with metabolic disease are at a high risk of cardiovascular complications ranging from microvascular dysfunction to cardiometabolic syndromes including heart failure. Despite significant advances in the standards of care for obese and diabetic patients, current therapeutic approaches are not always successful in averting the accompanying cardiovascular deterioration. There is a strong relationship between adipose inflammation seen in metabolic disorders and detrimental changes in cardiovascular structure and function. The particular importance of epicardial and perivascular adipose pools emerged as main modulators of the physiology or pathology of heart and blood vessels. Here, we review the peculiarities of these two fat depots in terms of their origin, function, and pathological changes during metabolic deterioration. We highlight the rationale for pharmacological targeting of the perivascular and epicardial adipose tissue or associated signaling pathways as potential disease modifying approaches in cardiometabolic syndromes.
Collapse
|
32
|
Bouras H, Roig SR, Kurstjens S, Tack CJJ, Kebieche M, de Baaij JHF, Hoenderop JGJ. Metformin regulates TRPM6, a potential explanation for magnesium imbalance in type 2 diabetes patients. Can J Physiol Pharmacol 2020; 98:400-411. [PMID: 32017603 DOI: 10.1139/cjpp-2019-0570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metformin therapy is associated with lower serum magnesium (Mg2+) levels in type 2 diabetes patients. The TRPM6 channel determines the fine-tuning of Mg2+ (re)absorption in intestine and kidney. Therefore, we aimed to investigate the short- and long-term effects of metformin on TRPM6. Patch clamp recordings and biotinylation assays were performed upon 1 h of incubation with metformin in TRPM6-transfected HEK293 cells. Additionally, 24 h of treatment of mDCT15 kidney and hCaco-2 colon cells with metformin was applied to measure the effects on endogenous TRPM6 expression by quantitative real-time PCR. To assess Mg2+ absorption, 25Mg2+ uptake measurements were performed using inductively coupled plasma mass spectrometry. Short-term effects of metformin significantly increased TRPM6 activity and its cell surface trafficking. In contrast, long-term effects significantly decreased TRPM6 mRNA expression and 25Mg2+ uptake. Metformin lowered TRPM6 mRNA levels independently of insulin- and AMPK-mediated pathways. Moreover, in type 2 diabetes patients, metformin therapy was associated with lower plasma Mg2+ concentrations and fractional excretion of Mg2+. Thereby, short-term metformin treatment increases TRPM6 activity explained by enhanced cell surface expression. Conversely, long-term metformin treatment results in downregulation of TRPM6 gene expression in intestine and kidney cells. This long-term effect translated in an inverse correlation between metformin and plasma Mg2+ concentration in type 2 diabetes patients.
Collapse
Affiliation(s)
- Hacene Bouras
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.,Faculty of Nature and Life Sciences, University of Mohamed Seddik Ben Yahia, Jijel, Algeria
| | - Sara R Roig
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Steef Kurstjens
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cees J J Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mohamed Kebieche
- Faculty of Nature and Life Sciences, University of Batna2, Algeria
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
33
|
Abstract
Metformin is a widely used biguanide drug due to its safety and low cost. It has been used for over 60 years to treat type 2 diabetes at the early stages because of its outstanding ability to decrease plasma glucose levels. Over time, different uses of metformin were discovered, and the benefits of metformin for various diseases and even aging were verified. These diseases include cancers (e.g., breast cancer, endometrial cancer, bone cancer, colorectal cancer, and melanoma), obesity, liver diseases, cardiovascular disease, and renal diseases. Metformin exerts different effects through different signaling pathways. However, the underlying mechanisms of these different benefits remain to be elucidated. The aim of this review is to provide a brief summary of the benefits of metformin and to discuss the possible underlying mechanisms.
Collapse
Affiliation(s)
- Ziquan Lv
- Department of Molecular Epidemiology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yajie Guo
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Yajie Guo
| |
Collapse
|
34
|
Elkhatib MAW, Mroueh A, Rafeh RW, Sleiman F, Fouad H, Saad EI, Fouda MA, Elgaddar O, Issa K, Eid AH, Eid AA, Abd-Elrahman KS, El-Yazbi AF. Amelioration of perivascular adipose inflammation reverses vascular dysfunction in a model of nonobese prediabetic metabolic challenge: potential role of antidiabetic drugs. Transl Res 2019; 214:121-143. [PMID: 31408626 DOI: 10.1016/j.trsl.2019.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
The onset of vascular impairment precedes that of diagnostic hyperglycemia in diabetic patients suggesting a vascular insult early in the course of metabolic dysfunction without a well-defined mechanism. Mounting evidence implicates adipose inflammation in the pathogenesis of insulin resistance and diabetes. It is not certain whether amelioration of adipose inflammation is sufficient to preclude vascular dysfunction in early stages of metabolic disease. Recent findings suggest that antidiabetic drugs, metformin, and pioglitazone, improve vascular function in prediabetic patients, without an indication if this protective effect is mediated by reduction of adipose inflammation. Here, we used a prediabetic rat model with delayed development of hyperglycemia to study the effect of metformin or pioglitazone on adipose inflammation and vascular function. At the end of the metabolic challenge, these rats were neither obese, hypertensive, nor hyperglycemic. However, they showed increased pressor responses to phenylephrine and augmented aortic and mesenteric contraction. Vascular tissues from prediabetic rats showed increased Rho-associated kinase activity causing enhanced calcium sensitization. An elevated level of reactive oxygen species was seen in aortic tissues together with increased Transforming growth factor β1 and Interleukin-1β expression. Although, no signs of systemic inflammation were detected, perivascular adipose inflammation was observed. Adipocyte hypertrophy, increased macrophage infiltration, and elevated Transforming growth factor β1 and Interleukin-1β mRNA levels were seen. Two-week treatment with metformin or pioglitazone or switching to normal chow ameliorated adipose inflammation and vascular dysfunction. Localized perivascular adipose inflammation is sufficient to trigger vascular dysfunction early in the course of diabetes. Interfering with this inflammatory process reverses this early abnormality.
Collapse
Affiliation(s)
- Mohammed A W Elkhatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ali Mroueh
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Rim W Rafeh
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Fatima Sleiman
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Hosny Fouad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Evan I Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohamed A Fouda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ola Elgaddar
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Khodr Issa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon; Department of Biomedical Sciences, Qatar University, Doha, Qatar
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon
| | - Khaled S Abd-Elrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa Brain and Mind Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
35
|
Green CJ, Marjot T, Tomlinson JW, Hodson L. Of mice and men: Is there a future for metformin in the treatment of hepatic steatosis? Diabetes Obes Metab 2019; 21:749-760. [PMID: 30456918 DOI: 10.1111/dom.13592] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver diseases, of which the first stage is steatosis. It is one of the most common liver diseases in developed countries and there is a clear association between type 2 diabetes (T2DM) and NAFLD. It is estimated that 70% of people with T2DM have NAFLD and yet there is currently no licensed pharmacological agent to treat it. Whilst lifestyle modification may ameliorate liver fat, it is often difficult to achieve or sustain; thus, there is great interest in pharmacological treatments for NAFLD. Metformin is the first-line medication in the management of T2DM and evidence from animal and human studies has suggested that it may be useful in reducing liver fat via inhibition of lipogenesis and increased fatty acid oxidation. Findings from the majority of studies undertaken in rodent models clearly suggest that metformin may be a powerful therapeutic agent specifically to reduce liver fat accumulation; data from human studies are less convincing. In the present review we discuss the evidence for the specific effects of metformin treatment on liver fat accumulation in animal and human studies, as well as the underlying proposed mechanisms, to try and understand and reconcile the difference in findings between rodent and human work in this area.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
36
|
Karise I, Bargut TC, Del Sol M, Aguila MB, Mandarim-de-Lacerda CA. Metformin enhances mitochondrial biogenesis and thermogenesis in brown adipocytes of mice. Biomed Pharmacother 2019; 111:1156-1165. [PMID: 30841429 DOI: 10.1016/j.biopha.2019.01.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/04/2019] [Accepted: 01/06/2019] [Indexed: 11/18/2022] Open
Abstract
AIMS We studied the effect of metformin on the brown adipose tissue (BAT) in a fructose-rich-fed model, focusing on BAT proliferation, differentiation, and thermogenic markers. MAIN METHODS C57Bl/6 mice received isoenergetic diets for ten weeks: control (C) or high-fructose (F). For additional eight weeks, animals received metformin hydrochloride (M, 250 mg/kg/day) or saline. After sacrifice, BAT and white fat pads were prepared for light microscopy and molecular analyses. KEY FINDINGS Body mass gain, white fat pads, and adiposity index were not different among the groups. There was a reduction in energy intake in the F group and energy expenditure in the F and FM groups. Metformin led to a more massive BAT in both groups CM and FM, associated with a higher adipocyte proliferation (β1-adrenergic receptor, proliferating cell nuclear antigen, and vascular endothelial growth factor), and differentiation (PR domain containing 16, bone morphogenetic protein 7), in part by activating 5' adenosine monophosphate-activated protein kinase. Metformin also enhanced thermogenic markers in the BAT (uncoupling protein type 1, peroxisome proliferator-activated receptor gamma coactivator-1 alpha) through adrenergic stimuli and fibroblast growth factor 21. Metformin might improve mitochondrial biogenesis in the BAT (nuclear respiratory factor 1, mitochondrial transcription factor A), lipolysis (perilipin, adipose triglyceride lipase, hormone-sensitive lipase), and fatty acid uptake (lipoprotein lipase, cluster of differentiation 36, adipocyte protein 2). SIGNIFICANCE Metformin effects are not linked to body mass changes, but affect BAT thermogenesis, mitochondrial biogenesis, and fatty acid uptake. Therefore, BAT may be a metformin adjuvant target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Iara Karise
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Thereza Cristina Bargut
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Mariano Del Sol
- Doctoral Program in Morphological Sciences, Universidad de La Frontera, Temuco, Chile.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
37
|
Yuan T, Li J, Zhao WG, Sun W, Liu SN, Liu Q, Fu Y, Shen ZF. Effects of metformin on metabolism of white and brown adipose tissue in obese C57BL/6J mice. Diabetol Metab Syndr 2019; 11:96. [PMID: 31788033 PMCID: PMC6880501 DOI: 10.1186/s13098-019-0490-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To investigate effects of metformin on the regulation of proteins of white adipose tissue (WAT) and brown adipose tissue (BAT) in obesity and explore the underlying mechanisms on energy metabolism. METHODS C57BL/6J mice were fed with normal diet (ND, n = 6) or high-fat diet (HFD, n = 12) for 22 weeks. HFD-induced obese mice were treated with metformin (MET, n = 6). After treatment for 8 weeks, oral glucose tolerance test (OGTT) and hyperinsulinemic-euglycemic clamp were performed to evaluate the improvement of glucose tolerance and insulin sensitivity. Protein expressions of WAT and BAT in mice among ND, HFD, and MET group were identified and quantified with isobaric tag for relative and absolute quantification (iTRAQ) coupled with 2D LC-MS/MS. The results were analyzed by MASCOT, Scaffold and IPA. RESULTS The glucose infusion rate in MET group was increased significantly compared with HFD group. We identified 4388 and 3486 proteins in WAT and BAT, respectively. As compared MET to HFD, differential expressed proteins in WAT and BAT were mainly assigned to the pathways of EIF2 signaling and mitochondrial dysfunction, respectively. In the pathways, CPT1a in WAT, CPT1b and CPT2 in BAT were down-regulated by metformin significantly. CONCLUSIONS Metformin improved the body weight and insulin sensitivity of obese mice. Meanwhile, metformin might ameliorate endoplasmic reticulum stress in WAT, and affect fatty acid metabolism in WAT and BAT. CPT1 might be a potential target of metformin in WAT and BAT.
Collapse
Affiliation(s)
- Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of The National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Juan Li
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei-Gang Zhao
- Department of Endocrinology, Key Laboratory of Endocrinology of The National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shuai-Nan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quan Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Fu
- Department of Endocrinology, Key Laboratory of Endocrinology of The National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhu-Fang Shen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Diabetes Research Center of Chinese Academy of Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Metformin: An Old Drug with New Applications. Int J Mol Sci 2018; 19:ijms19102863. [PMID: 30241400 PMCID: PMC6213209 DOI: 10.3390/ijms19102863] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022] Open
Abstract
Metformin is a biguanide drug that has been used to treat type 2 diabetes mellitus for more than 60 years. The United Kingdom Prospective Diabetic Study (UKPDS) has shown metformin to improve mortality rates in diabetes patients, and recent studies suggest metformin has additional effects in treating cancer, obesity, nonalcoholic fatty liver disease (NAFLD), polycystic ovary syndrome (PCOS), and metabolic syndrome. Metformin has also been shown to alleviate weight gain associated with antipsychotic medication. Metformin has recently been extensively studied and emerging evidence suggests metformin decreases hepatocyte triglyceride accumulation in NAFLD and prevents liver tumorigenesis. Interestingly, studies have also shown metformin reduces visceral fat, suppresses white-adipose-tissue (WAT) extracellular matrix remodeling, and inhibits obesity-induced inflammation. However, clinical evidence for using metformin to treat NAFLD, cancer, metabolic syndrome, or to prevent hepatocellular carcinoma in NAFLD patients is lacking. This review therefore addresses the potential beneficial effects of metformin on NAFLD, its role in protecting against cardiac ischemia–reperfusion (I/R) injury, atherosclerosis, glucotoxicity, and lipotoxicity induced oxidative and ER stress in pancreatic β-cell dysfunction, as well as its underlying molecular mechanisms of action.
Collapse
|