Category: Children

Metformin mechanism of action

Metformin mechanism of action

Factors associated with hospital admission mdchanism critical Merformin among Metformin mechanism of action with coronavirus disease in New Eye health supplements city: Mefformin cohort study. Metformin enhances mitochondrial biogenesis and thermogenesis in brown adipocytes of mice. G3 Bethesda. AMP-activated protein kinase signaling protects oligodendrocytes that restore central nervous system functions in an experimental autoimmune encephalomyelitis model. Article CAS Google Scholar Czyzyk, A.

Metformin mechanism of action -

Krall AS, Mullen PJ, Surjono F, et al. Asparagine couples mitochondrial respiration to ATF4 activity and tumor growth. Liu X, Chhipa RR, Pooya S, et al. Discrete mechanisms of mTOR and cell cycle regulation by AMPK agonists independent of AMPK. Yu X, Zhang Y, Xiong S, et al.

Gutkind JS, Molinolo AA, Wu X, et al. Inhibition of mTOR signaling and clinical activity of metformin in oral premalignant lesions. JCI Insight.

Mossmann D, Park S, Hall MN. mTOR signalling and cellular metabolism are mutual determinants in cancer. Yamaguchi H, Hsu JM, Yang WH, Hung MC. Mechanisms regulating PD-L1 expression in cancers and associated opportunities for novel small-molecule therapeutics.

Nat Rev Clin Oncol. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy.

Wei Z, Zhang X, Yong T, et al. Boosting anti-PD-1 therapy with metformin-loaded macrophage-derived microparticles. Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin.

Wabitsch S, McCallen JD, Kamenyeva O, et al. J Hepatol. Cha JH, Yang WH, Xia W, et al. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Munoz LE, Huang L, Bommireddy R, et al. Metformin reduces PD-L1 on tumor cells and enhances the anti-tumor immune response generated by vaccine immunotherapy.

J Immunother Cancer. Chung YM, Khan PP, Wang H, et al. Wen M, Cao Y, Wu B, et al. PD-L1 degradation is regulated by electrostatic membrane association of its cytoplasmic domain.

Yang Y, Hsu JM, Sun L, et al. Palmitoylation stabilizes PD-L1 to promote breast tumor growth. Cell Res. Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Hirsch HA, Iliopoulos D, Struhl K. Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth.

Cameron AR, Morrison VL, Levin D, et al. Anti-inflammatory effects of metformin irrespective of diabetes status. Pernicova I, Kelly S, Ajodha S, et al. Metformin to reduce metabolic complications and inflammation in patients on systemic glucocorticoid therapy: a randomised, double-blind, placebo-controlled, proof-of-concept, phase 2 trial.

Lancet Diabetes Endocrinol. Van Nostrand JL, Hellberg K, Luo EC, et al. AMPK regulation of Raptor and TSC2 mediate metformin effects on transcriptional control of anabolism and inflammation.

Genes Dev. Xian H, Liu Y, Rundberg Nilsson A, et al. Metformin inhibition of mitochondrial ATP and DNA synthesis abrogates NLRP3 inflammasome activation and pulmonary inflammation. Zhong Z, Liang S, Sanchez-Lopez E, et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation.

Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Martin-Montalvo A, Mercken EM, Mitchell SJ, et al. Metformin improves healthspan and lifespan in mice. Samaras K, Makkar S, Crawford JD, et al. Metformin use is associated with slowed cognitive decline and reduced incident dementia in older adults with type 2 diabetes: the Sydney memory and ageing study.

Diabetes Care. Charpignon ML, Vakulenko-Lagun B, Zheng B, et al. Causal inference in medical records and complementary systems pharmacology for metformin drug repurposing towards dementia. Bharath LP, Agrawal M, McCambridge G, et al. Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation.

Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. Apolzan JW, Venditti EM, Edelstein SL, et al.

Long-term weight loss with metformin or lifestyle intervention in the diabetes prevention program outcomes study. Ann Intern Med. Kahn SE, Haffner SM, Heise MA, et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy.

N Engl J Med. Diabetes Prevention Program Research Group. Long-term safety, tolerability, and weight loss associated with metformin in the diabetes prevention program outcomes study.

Day EA, Ford RJ, Smith BK, et al. Metformin-induced increases in GDF15 are important for suppressing appetite and promoting weight loss. Nat Metab. Coll AP, Chen M, Taskar P, et al. GDF15 mediates the effects of metformin on body weight and energy balance.

Wang D, Day EA, Townsend LK, Djordjevic D, Jorgensen SB, Steinberg GR. GDF emerging biology and therapeutic applications for obesity and cardiometabolic disease. Mullican SE, Lin-Schmidt X, Chin CN, et al.

GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates. Gerstein HC, Pare G, Hess S, et al. Growth differentiation factor 15 as a novel biomarker for metformin. Download references. The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Department of Physiology and Pathophysiology, Jerry M. Wallace School of Osteopathic Medicine, Campbell University SOM, Buies Creek, NC, USA.

Department of Biology, College of Arts and Sciences, University of North Carolina, Greensboro, NC, USA. Department of Pharmacology, Jerry M. Wallace School of Osteopathic Medicine, Campbell University, Buies Creek, NC, USA. Geisinger Commonwealth School of Medicine, Scranton, PA, USA.

You can also search for this author in PubMed Google Scholar. All authors contributed to the review article topic conception, literature searching and analysis, and the writing of the manuscript. All authors read and approved the final manuscript.

Correspondence to Hong Zhu. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Springer Nature or its licensor e. a society or other partner holds exclusive rights to this article under a publishing agreement with the author s or other rightsholder s ; author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions. Zhu, H. et al. Molecular mechanisms of action of metformin: latest advances and therapeutic implications. Clin Exp Med 23 , — Download citation. Received : 26 February Accepted : 17 March Published : 04 April Issue Date : November Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Download PDF. Abstract Metformin is among the most widely used antidiabetic drugs.

Glucose transporters in adipose tissue, liver, and skeletal muscle in metabolic health and disease Article Open access 26 June Antiaging agents: safe interventions to slow aging and healthy life span extension Article Open access 09 May Use our pre-submission checklist Avoid common mistakes on your manuscript.

Overview Metformin 1,1-dimethylbiguanide structure in Fig. Chemical structure of metformin 1,2-dimethylbiguanide. Full size image.

Novel molecular targets involved in glycemic control Inhibition of hepatic gluconeogenesis is a major action of metformin in lowering blood glucose. AMPK activation How metformin activates AMPK remains unclear. MicroRNA modulation, another AMPK-independent mechanism The evolutionarily conserved transcription factor hepatocyte nuclear factor 4 alpha HNF4α plays an important role in promoting hepatic glucose production and regulating energy balance [ 13 ].

Novel molecular targets involved in cardiovascular protection Extensive studies in animal models suggested an important role for metformin in protecting against diverse cardiovascular disorders, including atherosclerosis, hypertension, myocardial ischemic injury, and heart failure reviewed in [ 2 ] , as well as cardiovascular toxicity induced by drugs and environmental pollutants [ 19 , 20 ].

Novel molecular targets involved in anticancer activities Direct and indirect mechanisms Numerous studies in animal models and multiple cohort studies in humans show an anticancer activity for metformin reviewed in [ 2 , 3 ].

Anti-inflammation The anti-inflammatory activities of metformin have been observed in various animal models and human subjects [ 58 , 59 , 60 ]. Antiaging activity Aging is characterized by a progressive loss of physiological integrity, leading to impaired function and increased vulnerability to developing chronic diseases, such as cardiovascular disorders, neurodegenerative diseases, and some types of cancers, among others.

Weight control and energy balance It is well established that metformin treatment is associated with weight loss in diabetic and non-diabetic people [ 70 , 71 , 72 ]. Conclusion and perspectives In conclusion, multiple novel molecular targets of metformin action have been discovered over the past few years Fig.

Boosting GLP-1 by Natural Products Chapter © Data availability Not applicable since this is a review article. Abbreviations AMPK: Adenosine monophosphate-activated kinase ERAD: Endoplasmic reticulum-associated protein degradation GDF Growth differentiation factor 15 GPD2: Mitochondrial glycerolphosphate dehydrogenase HNF4α: Hepatocyte nuclear factor 4 alpha IL: Interleukin METC: Mitochondrial electron transport chain mtDNA: Mitochondrial DNA mTOR: Mammalian target of rapamycin ORAC: Calcium release-activated channel ox-mtDNA: Oxidized mitochondrial DNA PM: Particulate matter ROS: Reactive oxygen species.

References Flory J, Lipska K. Article PubMed PubMed Central Google Scholar Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Article CAS PubMed Google Scholar Vasan K, Werner M, Chandel NS. Article CAS PubMed PubMed Central Google Scholar Kulkarni AS, Gubbi S, Barzilai N.

Article CAS PubMed PubMed Central Google Scholar He L, Wondisford FE. Article CAS PubMed Google Scholar Paleari L, Burhenne J, Weiss J, et al. Article PubMed Google Scholar Zake DM, Kurlovics J, Zaharenko L, Komasilovs V, Klovins J, Stalidzans E.

Article CAS PubMed PubMed Central Google Scholar Ma T, Tian X, Zhang B, et al. Article CAS PubMed PubMed Central Google Scholar Duca FA, Cote CD, Rasmussen BA, et al. Article CAS PubMed PubMed Central Google Scholar Madiraju AK, Erion DM, Rahimi Y, et al. Article CAS PubMed PubMed Central Google Scholar Madiraju AK, Qiu Y, Perry RJ, et al.

Article CAS PubMed PubMed Central Google Scholar LaMoia TE, Butrico GM, Kalpage HA, et al. Article CAS PubMed PubMed Central Google Scholar Girard R, Tremblay S, Noll C, et al.

Article CAS PubMed PubMed Central Google Scholar Da L, Cao T, Sun X, et al. Article CAS Google Scholar Xie D, Chen F, Zhang Y, et al. Article CAS PubMed PubMed Central Google Scholar Howell JJ, Hellberg K, Turner M, et al.

Article CAS PubMed Google Scholar Sabatini DM. Article CAS PubMed PubMed Central Google Scholar Waise TMZ, Rasti M, Duca FA, et al. Article CAS PubMed PubMed Central Google Scholar Soberanes S, Misharin AV, Jairaman A, et al.

Article CAS PubMed Google Scholar Efentakis P, Kremastiotis G, Varela A, et al. Article CAS PubMed Google Scholar Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes UKPDS Article CAS PubMed PubMed Central Google Scholar Larsen AH, Jessen N, Norrelund H, et al.

Article CAS PubMed Google Scholar Salt IP, Hardie DG. Article CAS PubMed PubMed Central Google Scholar Winnik S, Auwerx J, Sinclair DA, Matter CM. Article CAS PubMed PubMed Central Google Scholar Tang X, Chen XF, Wang NY, et al. Article CAS PubMed PubMed Central Google Scholar Lai YC, Tabima DM, Dube JJ, et al.

Article CAS PubMed PubMed Central Google Scholar Gao P, You M, Li L, et al. Article CAS PubMed Google Scholar Mizushima N, Komatsu M. Article CAS PubMed Google Scholar Bravo-San Pedro JM, Kroemer G, Galluzzi L.

Article CAS PubMed Google Scholar Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer G. Article CAS PubMed Google Scholar Xie Z, Lau K, Eby B, et al.

Article CAS PubMed PubMed Central Google Scholar Vasamsetti SB, Karnewar S, Kanugula AK, Thatipalli AR, Kumar JM, Kotamraju S.

Article CAS PubMed Google Scholar Seneviratne A, Cave L, Hyde G, et al. In the longitudinal study UKPDS [ 3 ], it was found that taking metformin significantly reduces mortality from all causes.

Studies in non-diabetic rats has confirmed that metformin improves heart function and reduces the size of lesions after induction of myocardial infarction [ 24 ]. This beneficial effect is the result of a myocardial preconditioning induced by metformin with varying degrees of permanence. One fundamental effect of metformin is to induce metabolic adaptation of the myocardium in critical energy situations such as ischaemia.

In a rodent model of heart failure, metformin promotes metabolic adaptation in the heart by stimulating gradual consumption of glucose instead of fatty acids. This action appears to be dependent upon activation of AMPK, which contributes to stimulating glucose uptake and glycolysis activity to cope with oxygen deprivation [ 25 ].

Metformin has been found to have beneficial effects in diabetic patients with a history of heart failure, reducing both mortality and morbidity [ 26 ]. One of the mechanisms thought to be behind diabetic cardiomyopathy and heart failure is defective autophagy in the heart. Metformin restores autophagy in diabetic mice and prevents the formation of cardiac lesions.

AMPK must be activated, since metformin is ineffective in diabetic mice with defective AMPK activity in their hearts [ 27 ]. The cardioprotective effects of metformin stem from more than one mechanism and both macro- and microvascular systems are involved.

Metformin is known to exercise a protective effect on the vascular endothelium, both by decreasing the production of free radicals and by reducing the formation of glycated proteins, which cause oxidative stress and inflammation.

The first studies suggesting a possible role for metformin in oncology were published in [ 28 ]. Since then, many other observational epidemiological studies have confirmed that type 2 diabetic patients who have been taking metformin for several years are at lower risk of developing or dying from cancer [ 29 ].

A relationship has also been found between the dose of metformin or duration of treatment and the protection observed, giving further evidence of anticancer properties [ 28 ].

A wide variety of mechanisms has been described to explain its beneficial effects Figure 3 , as they are dependent upon the metabolic and molecular characteristics of each type of tumour. Figure 3. Several different modes of action have been suggested, due to the diversity of in vivo and in vitro cancer models studied.

At cell level, metformin inhibits the mTORC1 mammalian target of rapamycin complex 1 pathway via mechanisms dependent on AMPK activation by phosphorylation of TSC2 tuberous sclerosis complex 2 and raptor regulatory associated protein of mTOR. However, metformin is able to inhibit the mTORC1 pathway via AMPK-independent mechanisms, by inhibiting Rag GTPases and inducing REDD1 expression.

It is possible that the drug acts at the level of cell cycle regulation by inhibiting cyclin D1 expression. Metformin causes a reduction in neoplastic cell angiogenesis by lowering blood concentrations of PAI-1 plasminogen activator inhibitor-1 and VEGF vascular endothelial growth factor.

Inhibition of metastasis formation through decreased activity of the metalloproteinases MMP2 and -9 has also been described.

Several epidemiological studies have reported a strong link between type 2 diabetes and elevated risk of certain types of cancer, in particular colon, endometrium, rectum and breast cancer, compared with the incidence of these cancers in the non-diabetic population.

It is thought that compensatory hyperinsulinaemia and chronic hyperglycaemia, both characteristics of type 2 diabetes, are behind this link between diabetes and cancer.

Insulin is known to promote cell proliferation and increase blood levels of IGF1 insulin-like growth factor 1 , which acts as a growth factor in tumour development. It is also well established that glucose is the preferred energy substrate used by cells during proliferation and hyperglycaemia could therefore contribute to increasing the growth and survival of cancer cells.

The inhibition of tumour progression by metformin has been confirmed through studies of large numbers of mouse carcinogenesis models, as well as the observational epidemiological studies. In some models, the inhibition of tumour proliferation was directly linked to reduction in blood insulin and IGF1 levels by metformin Figure 3 and reduced activity of receptors with tyrosine kinase activity [ 30 ].

However, this effect was not observed in other models, suggesting a mode of action working independently of decreased blood insulin levels. Several clinical trials have suggested that metformin is able to procure a protective effect against cancer regardless of whether the patient has diabetes [ 31 ].

This finding has been supported by a large body of in vitro evidence demonstrating that the drug directly inhibits the proliferation of cancer cells [ 32 , 33 ].

It is important to point out that tumour cells have varying degrees of sensitivity to metformin and its antiproliferative effect is only observed at concentrations much higher mM than those used to treat diabetes plasma concentrations in the region of 10—40 μM.

Nonetheless, metformin does accumulate in some organs, such as the intestine and liver, allowing much higher intracellular concentrations than those in the plasma. Moreover, in a carcinogenesis model induced via the loss of the tumour suppressor PTEN phosphatase and tensin homolog , activation of AMPK by a direct activator A produces a reduction in tumour progression identical to that obtained with metformin [ 34 ].

AMPK directly phosphorylates the tumour suppressor TSC2 tuberous sclerosis complex 2 and the regulator protein raptor regulatory associated protein of mTOR , leading to rapid suppression of mTORC1 signalling pathway Figure 3. This inhibition of the mTORC1 pathway by metformin leads to decreased expression of EGF epidermal growth factor receptor and the oncoprotein HER2 erbB-2 in breast cancer lines.

A study of gene expression profiles in breast cancer tumours treated with metformin revealed a reduction in the expression of genes involved in mitosis progression.

The metabolism of cancer cells is essentially based on consuming glucose to produce energy, even when oxygen is present. This feature means that cancer cells preferentially use glycolysis in aerobic conditions Warburg effect [ 52 ] or aerobic glycolysis , as opposed to mitochondrial oxidative phosphorylation.

However, cancer cells use their mitochondria to produce some metabolites malate, citrate needed for the various biosynthetic pathways required for cell proliferation. By blocking the respiratory chain, metformin will therefore inhibit this adaptive metabolism and cause a major energy crisis.

For example, tumour cells with a loss of p53 function cannot cope with the metabolic changes imposed by metformin which require activation of p53 by AMPK and die from apoptosis [ 36 ].

Similarly, cancer cells that are deficient in LKB1 showing low AMPK activity are more sensitive to the ATP depletion imposed by metformin because of their inability to restore their energy balance via AMPK activation.

In these situations, metformin acts on cancer cells as a selective cytotoxic agent. Changes in the tumour microenvironment may also contribute to the beneficial effects of metformin. The possibility has also been raised that it may inhibit tumour angiogenesis via a reduction in blood levels of PAI-1 plasminogen activator inhibitor-1 and VEGF vascular endothelial growth factor Figure 3.

However, the opposite was found in a different study, reporting an increase in intratumoural microvascular density in tumour xenograft cells [ 31 ].

Metformin participates in the response to damaged DNA by selectively activating ATM ataxia telangiectasia mutated , but also by inhibiting reactive oxygen species production brought about through transformation by the oncogene Ras [ 38 ].

Metformin also contributes to preventing tumour development by controlling cancer stem cell ontogenesis, as it targets the epithelial-mesenchymal transition and differentiation in these cells [ 39 , 40 ]. This specific action of metformin on cancer stem cells has opened up new possibilities in cancer prevention, due to the resistance of these cells to chemotherapy.

It is significant that metformin treatment has been linked to a better response rate to chemotherapy for breast cancer [ 41 ]. This synergistic effect of metformin on the cytotoxicity of chemotherapy agents has been confirmed in vitro and has been found to be effective in reducing resistance to these drugs, which supports the potential use of metformin as a chemotherapy adjuvant [ 42 ].

Lastly, recent studies have suggested a role for metformin in inhibiting the invasive potential of cancer cells and the formation of metastasis by decreasing the activity of the metalloproteinases MMP2 and MMP9 [ 40 , 43 ] Figure 3. However, it is now important to identify which patients might benefit from its potential for preventing and treating cancer, by investigating biomarkers that will predict its therapeutic effect.

The sensitivity of cancer cells to metformin depends on the type of cancer, the presence of mutations for example for LKB1, p53 or OCT1 polymorphisms and the tumour environment. On the basis of the pharmacokinetic properties of phenformin membrane permeability, potent complex I inhibition , several authors have suggested that this drug previously taken off the market could be used as a much more effective antineoplastic agent than metformin [ 34 ].

Future studies will need to determine the optimal tolerable doses of both metformin and phenformin for the treatment of different cancers. Over one hundred phase II and III clinical trials are currently under way to assess the use of metformin in oncology 2.

Recent studies have opened up the prospect of possible therapeutic indications for metformin in the treatment of neurodegenerative diseases. Metformin induces dephosphorylation of tau by activating the protein phosphatase 2A PP2A , and thus slows the progression of the disease.

Another recent study also showed that metformin promotes neurogenesis and enhances hippocampus-dependent spatial memory formation and learning [ 48 ]. Metformin promotes phosphorylation of CBP CREB binding protein , a transcription coactivator with intrinsic histone acetyltransferase activity, via the protein kinase C PKC -ζ.

This stage is crucial for the differentiation of radial glial neuronal precursors and the formation of new neurons in the hippocampus. The protection afforded by metformin against diabetes, cardiovascular disease and cancer is similar to the anti-ageing effects of calorie restriction Figure 4.

There have been reports of metformin having properties that mimic calorie restriction and prolong the lifespan of the nematode Caenorhabditis elegans and rodents.

Figure 4. Summary of the principal effects of metformin. The principal antidiabetic effects of metformin occur in the liver, via inhibition of gluconeogenesis, and to a lesser extent in the intestine and muscle, leading to a reduction in hyperglycaemia and blood lipids and to increased insulin sensitivity.

These same improvements indirectly procure both cardiovascular protective and anticancer effects. Added to this, metformin directly reduces the risk of cardiovascular disease through actions that affect both macro- and microvascular systems and exerts a direct antitumoural effect on cancer cells.

Furthermore, metformin is thought to have a neuroprotective effect in neurodegenerative diseases. The beneficial effects of metformin are similar to those observed with calorie restriction, which help to prolong lifespan. Metformin can therefore be considered a calorie restriction mimetic with possible anti-ageing properties.

In non-diabetic mice, metformin mimics the effects of calorie restriction on the gene expression profile and prevents the onset of diabetes, cardiovascular disease and cancer [ 49 ]. It has recently been suggested that metformin indirectly increases the lifespan of nematodes by disrupting the metabolism of its accompanying microbe Escherichia coli [ 51 ].

At present, metformin is the most frequently used antidiabetic agent in the treatment of type 2 diabetes. Its success is due to various factors: efficacy, safety, good tolerability and low production cost. The drug also has the advantage of counteracting the cardiovascular complications associated with diabetes, by inducing myocardial preconditioning.

Another significant benefit of metformin is its demonstrated capacity to reduce the risk of tumour development by controlling cell differentiation and proliferation. New therapeutic indications may also be found for the drug in the treatment of neurodegenerative diseases.

This old drug may well have more surprises in store. The authors declare that they have no interests associated with the information published in this article.

Current usage metrics show cumulative count of Article Views full-text article views including HTML views, PDF and ePub downloads, according to the available data and Abstracts Views on Vision4Press platform.

Data correspond to usage on the plateform after The current usage metrics is available hours after online publication and is updated daily on week days. Free Access. Issue Med Sci Paris. Effect on longevity: metformin as an elixir of life? Conclusion Declaration of interests References List of figures.

Bailey C, Campbell I. Metformin: the gold standard; a scientific handbook Chichester, UK: Wiley, p. Du nouveau dans les antidiabétiques. La NN diméthylamino guanyl guanidine N. Maroc Med ; — Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes UKPDS Lancet ; — Metformin and lactic acidosis: cause or coincidence?

A review of case reports. J Intern Med ; — Metformin decreases food consumption and induces weight loss in subjects with obesity with type II non-insulin-dependent diabetes.

Obes Res ; 6: 47— Metformin reverses fatty liver disease in obese, leptin-deficient mice. Nat Med ; 6: — Metformin might influence tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction of energetic stress; however, these effects require further investigation.

Here, we discuss the updated understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries of metformin targets for the treatment of diabetes mellitus and cancer.

Abstract Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the mechanistic aspects of metformin action remained ill-defined. Publication types Research Support, Non-U.

Metformin Metformin mechanism of action among Metforimn most widely used antidiabetic Metformin mechanism of action. Actiion over the Metformin mechanism of action few years have identified multiple novel molecular targets and pathways that metformin acts on to Metcormin its beneficial effects in treating type 2 diabetes as Immune system modulation as Metforkin disorders involving dysregulated inflammation and redox homeostasis. In this mini-review, we discuss the latest cutting-edge research discoveries on novel molecular targets of metformin in glycemic control, cardiovascular protection, cancer intervention, anti-inflammation, antiaging, and weight control. Identification of these novel targets and pathways not only deepens our understanding of the molecular mechanisms by which metformin exerts diverse beneficial biological effects, but also provides opportunities for developing new mechanistically based drugs for human diseases. Zhe Li, Zhenkun Zhang, … Fangxia Guan. Actipn has been Antioxidant-rich beverages for detoxification first-line drug treatment for qction and insulin resistance for Metformin mechanism of action 50 years. However, Kidney bean wraps molecular basis adtion its therapeutic Pre-workout supplements remained incompletely understood. In addition, epidemiological and clinical observation studies suggest that metformin reduced cancer risk in patients with type actkon diabetes and improved survival outcome of human cancers. Experimental studies have shown that this drug can inhibit cancer cell viability, growth, and proliferation through inhibiting mTORC1 signaling and mitochondrial complex I, suggesting that it may be a promising drug candidate for malignancy. Here, we summarize recent progress in studies of metformin in type 2 diabetes and tumorigenesis, which provides novel insight on the therapeutic development of human diseases. Metformin, a derivative of guanidine, has been used in the treatment of hyperglycemia and type 2 diabetes mellitus T2DM for over 50 years American Diabetes Association,

Metformin mechanism of action has been the mainstay Metformin mechanism of action therapy for diabetes mellitus for many years; o, the mechanistic aspects of metformin action remained Metformib.

Recent advances revealed that this Metfoormin, in addition Anthocyanins in red wine its glucose-lowering Metfogmin, might be promising Caffeine pills for improved physical performance specifically Metformin mechanism of action metabolic differences between Metvormin and mechaniwm metabolic signalling.

The knowledge gained from dissecting the principal Fasting and longevity by eMtformin metformin works can help us mecbanism develop novel Metfomrin.

The centre of metformin's Metformin mechanism of action of mechajism is jechanism alteration sction the energy metabolism of the cell.

Metformin exerts its prevailing, glucose-lowering effect by inhibiting hepatic gluconeogenesis and opposing the action of glucagon. The inhibition of mitochondrial complex I results in defective cAMP and protein kinase A signalling in response to glucagon.

Stimulation of 5'-AMP-activated protein kinase, although dispensable for the glucose-lowering effect of metformin, confers insulin sensitivity, mainly by modulating lipid metabolism. Metformin might influence tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction of energetic stress; however, these effects require further investigation.

Here, we discuss the updated understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries of metformin targets for the treatment of diabetes mellitus and cancer. Abstract Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the mechanistic aspects of metformin action remained ill-defined.

Publication types Research Support, Non-U. Gov't Review. Substances Glucagon Metformin AMP-Activated Protein Kinases.

: Metformin mechanism of action

Publication types elegans and D. Twenty-five years of mTOR: uncovering the link from nutrients to growth. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Article CAS PubMed PubMed Central Google Scholar Vasamsetti SB, Karnewar S, Kanugula AK, Thatipalli AR, Kumar JM, Kotamraju S. Regular insulin. The demonstration is clear and straightforward, and the results may have a great impact not only on our understanding of metformin mechanism in humans but also on future metformin therapy in clinic, for example, using gut-released metformin Met DR instead of the current formulation Met XR.
Frontiers | Molecular Mechanisms of Metformin for Diabetes and Cancer Treatment

Stimulation of 5'-AMP-activated protein kinase, although dispensable for the glucose-lowering effect of metformin, confers insulin sensitivity, mainly by modulating lipid metabolism. Metformin might influence tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction of energetic stress; however, these effects require further investigation.

Here, we discuss the updated understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries of metformin targets for the treatment of diabetes mellitus and cancer.

Abstract Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the mechanistic aspects of metformin action remained ill-defined. Sterne J. Blood sugar-lowering effect of 1,1-dimethylbiguanide. Knaani Z , Klajman A. Our experiences with N,N-dimethyl-guanyl-guanidine La in the treatment of diabetic patients.

Bischoff F , Long ML. Guanidine structure and hypoglycemia. J Pharmacol Exp Ther. McKendry JB , Kuwayti K , Rado PP. Clinical experience with DBI phenformin in the management of diabetes. Can Med Assoc J. Ungar G , Freedman L , Shapiro SL. Pharmacological studies of a new oral hypoglycemic drug.

Proc Soc Exp Biol Med. Wien Med Wochenschr. Hermann LS. Metformin: a review of its pharmacological properties and therapeutic use.

Diabete Metab. Schäfer G. A review of history, pharmacodynamics and therapy. Reaven GM. Role of insulin resistance in human disease. Bailey CJ , Puah JA.

Effect of metformin on glucose metabolism in mouse soleus muscle. Jackson RA , Hawa MI , Jaspan JB , et al. Mechanism of metformin action in non-insulin-dependent diabetes.

Pagano G , Tagliaferro V , Carta Q , et al. Metformin reduces insulin requirement in Type 1 insulin-dependent diabetes. Pedersen O , Nielsen O , Bak J , Richelsen B , Beck-Nielsen H , Sørensen N. The effects of metformin on adipocyte insulin action and metabolic control in obese subjects with type 2 diabetes.

Diabet Med. Gin H , Messerchmitt C , Brottier E , Aubertin J. Metformin improved insulin resistance in type I, insulin-dependent, diabetic patients. World Health Organization. WHO model list of essential medicines. World Health Organization ; Accessed April 30, Evans JM , Donnelly LA , Emslie-Smith AM , Alessi DR , Morris AD.

Metformin and reduced risk of cancer in diabetic patients. Zakikhani M , Dowling R , Fantus IG , Sonenberg N , Pollak M. Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells.

Cancer Res. Barzilai N , Crandall JP , Kritchevsky SB , Espeland MA. Metformin as a Tool to Target Aging. Cell Metab. Justice JN , Ferrucci L , Newman AB , et al. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup.

Bharath LP , Agrawal M , McCambridge G , et al. Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell Metab ; 32 : 44 - Proc Natl Acad Sci U S A. Petrie JR , Chaturvedi N , Ford I , et al.

Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes REMOVAL : a double-blind, randomised, placebo-controlled trial.

Lancet Diabetes Endocrinol. Maruthur NM , Tseng E , Hutfless S , et al. Diabetes medications as monotherapy or metformin-based combination therapy for type 2 diabetes: a systematic review and meta-analysis.

Ann Intern Med. Grant PJ. The effects of metformin on cardiovascular risk factors. Diabetes Metab Rev. Juhan-Vague I , Vague P , Alessi MC , et al. Relationships between plasma insulin triglyceride, body mass index, and plasminogen activator inhibitor 1. Rains SG , Wilson GA , Richmond W , Elkeles RS.

The reduction of low density lipoprotein cholesterol by metformin is maintained with long-term therapy. J R Soc Med. McCreight LJ , Mari A , Coppin L , Jackson N , Umpleby AM , Pearson ER. Metformin increases fasting glucose clearance and endogenous glucose production in non-diabetic individuals.

Galuska D , Nolte LA , Zierath JR , Wallberg-Henriksson H. Effect of metformin on insulin-stimulated glucose transport in isolated skeletal muscle obtained from patients with NIDDM.

Inzucchi SE , Maggs DG , Spollett GR , et al. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. Stumvoll M , Nurjhan N , Perriello G , Dailey G , Gerich JE. Metabolic effects of metformin in non-insulin-dependent diabetes mellitus.

Samuel Varman T , Shulman Gerald I. Mechanisms for insulin resistance: common threads and missing links. Magnusson I , Rothman DL , Katz LD , Shulman RG , Shulman GI. Increased rate of gluconeogenesis in type II diabetes mellitus.

J Clin Invest. Nosadini R , Avogaro A , Trevisan R , et al. Effect of metformin on insulin-stimulated glucose turnover and insulin binding to receptors in type II diabetes. Johnson AB , Webster JM , Sum CF , et al. The impact of metformin therapy on hepatic glucose production and skeletal muscle glycogen synthase activity in overweight type II diabetic patients.

Hundal RS , Krssak M , Dufour S , et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Widén EI , Eriksson JG , Groop LC. Metformin normalizes nonoxidative glucose metabolism in insulin-resistant normoglycemic first-degree relatives of patients with NIDDM.

Forslund K , Hildebrand F , Nielsen T , et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Wu H , Esteve E , Tremaroli V , et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug.

Nat Med. Graham GG , Punt J , Arora M , et al. Clinical pharmacokinetics of metformin. Clin Pharmacokinet. Timmins P , Donahue S , Meeker J , Marathe P. Steady-state pharmacokinetics of a novel extended-release metformin formulation. Madiraju AK , Qiu Y , Perry RJ , et al. Metformin inhibits gluconeogenesis via a redox-dependent mechanism in vivo.

Kajbaf F , De Broe ME , Lalau JD. Therapeutic concentrations of metformin: a systematic review. Madiraju AK , Erion DM , Rahimi Y , et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Frid A , Sterner GN , Löndahl M , et al.

Novel assay of metformin levels in patients with type 2 diabetes and varying levels of renal function: clinical recommendations. Wang Y , Nasiri AR , Damsky WE , et al. Uncoupling hepatic oxidative phosphorylation reduces tumor growth in two murine models of colon cancer.

Cell Rep. Chandel NS , Avizonis D , Reczek CR , et al. Are metformin doses used in murine cancer models clinically relevant? Wilcock C , Bailey CJ. Accumulation of metformin by tissues of the normal and diabetic mouse. He L , Wondisford FE. Metformin action: concentrations matter.

Gormsen LC , Sundelin EI , Jensen JB , et al. In vivo imaging of human 11 C-Metformin in peripheral organs: dosimetry, biodistribution, and kinetic analyses. J Nucl Med. Owen MR , Doran E , Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain.

Biochem J. El-Mir MY , Nogueira V , Fontaine E , Avéret N , Rigoulet M , Leverve X. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. Foretz M , Hébrard S , Leclerc J , et al.

Drahota Z , Palenickova E , Endlicher R , et al. Biguanides inhibit complex I, II and IV of rat liver mitochondria and modify their functional properties. Physiol Res. Hunter RW , Hughey CC , Lantier L , et al.

Metformin reduces liver glucose production by inhibition of fructosebisphosphatase. He L , Sabet A , Djedjos S , et al. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein.

Bridges Hannah R , Jones Andrew JY , Pollak Michael N , Hirst J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Wang DS , Jonker JW , Kato Y , Kusuhara H , Schinkel AH , Sugiyama Y. Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin.

Jakobsen S , Busk M , Jensen JB , et al. A PET tracer for renal organic cation transporters, 11C-metformin: radiosynthesis and preclinical proof-of-concept studies.

Shu Y , Sheardown SA , Brown C , et al. Effect of genetic variation in the organic cation transporter 1 OCT1 on metformin action. Kwon EY , Chung JY , Park HJ , Kim BM , Kim M , Choi JH.

OCT3 promoter haplotype is associated with metformin pharmacokinetics in Koreans. Sci Rep. Tzvetkov MV , Vormfelde SV , Balen D , et al. The effects of genetic polymorphisms in the organic cation transporters OCT1, OCT2, and OCT3 on the renal clearance of metformin.

Clin Pharmacol Ther. Cusi K , Consoli A , DeFronzo RA. Metabolic effects of metformin on glucose and lactate metabolism in noninsulin-dependent diabetes mellitus.

J Clin Endocrinol Metab. Gormsen LC , Søndergaard E , Christensen NL , Brøsen K , Jessen N , Nielsen S. Metformin increases endogenous glucose production in non-diabetic individuals and individuals with recent-onset type 2 diabetes. Konopka AR , Esponda RR , Robinson MM , et al.

Hyperglucagonemia mitigates the effect of metformin on glucose production in prediabetes. Musi N , Hirshman MF , Nygren J , et al. Metformin increases AMP-activated protein kinase activity in skeletal muscle of subjects with type 2 diabetes.

Rossetti L , Smith D , Shulman GI , Papachristou D , DeFronzo RA. Correction of hyperglycemia with phlorizin normalizes tissue sensitivity to insulin in diabetic rats.

Yu JG , Kruszynska YT , Mulford MI , Olefsky JM. A comparison of troglitazone and metformin on insulin requirements in euglycemic intensively insulin-treated type 2 diabetic patients. Karlsson HKR , Hällsten K , Björnholm M , et al. Effects of metformin and rosiglitazone treatment on insulin signaling and glucose uptake in patients with newly diagnosed type 2 diabetes.

Dandona P , Fonseca V , Mier A , Beckett AG. Diarrhea and metformin in a diabetic clinic. Florez H , Luo J , Castillo-Florez S , et al.

Impact of metformin-induced gastrointestinal symptoms on quality of life and adherence in patients with type 2 diabetes. Postgrad Med. Dujic T , Zhou K , Donnelly LA , Tavendale R , Palmer CN , Pearson ER.

Association of organic cation transporter 1 with intolerance to metformin in type 2 diabetes: a GoDARTS study. McCreight LJ , Bailey CJ , Pearson ER. Metformin and the gastrointestinal tract.

Bailey CJ , Wilcock C , Scarpello JH. Metformin and the intestine. Bailey CJ , Mynett KJ , Page T. Importance of the intestine as a site of metformin-stimulated glucose utilization. Br J Pharmacol. Coll AP , Chen M , Taskar P , et al. GDF15 mediates the effects of metformin on body weight and energy balance.

Lee A , Morley JE. Metformin decreases food consumption and induces weight loss in subjects with obesity with type II non-insulin-dependent diabetes. Obes Res. Day EA , Ford RJ , Smith BK , et al. Metformin-induced increases in GDF15 are important for suppressing appetite and promoting weight loss.

Nat Metab. Natali A , Nesti L , Venturi E , et al. Metformin is the key factor in elevated plasma growth differentiation factor levels in type 2 diabetes: a nested, case—control study. Diabetes Obes Metab. Gontier E , Fourme E , Wartski M , et al. High and typical 18F-FDG bowel uptake in patients treated with metformin.

Eur J Nucl Med Mol Imaging. Migoya EM , Bergeron R , Miller JL , et al. Dipeptidyl peptidase-4 inhibitors administered in combination with metformin result in an additive increase in the plasma concentration of active GLP Wu T , Thazhath SS , Bound MJ , Jones KL , Horowitz M , Rayner CK.

Mechanism of increase in plasma intact GLP-1 by metformin in type 2 diabetes: stimulation of GLP-1 secretion or reduction in plasma DPP-4 activity? Diabetes Res Clin Pract. Ekberg K , Landau BR , Wajngot A , et al. Contributions by kidney and liver to glucose production in the postabsorptive state and after 60 h of fasting.

Rothman DL , Magnusson I , Katz LD , Shulman RG , Shulman GI. Quantitation of hepatic glycogenolysis and gluconeogenesis in fasting humans with 13 C NMR. Lin HV , Accili D. Hormonal regulation of hepatic glucose production in health and disease. Petersen MC , Vatner DF , Shulman GI. Regulation of hepatic glucose metabolism in health and disease.

Nat Rev Endocrinol. Insulin regulates liver metabolism in vivo in the absence of hepatic Akt and Foxo1. Perry RJ , Camporez JG , Kursawe R , et al. Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes.

Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver. Samuel VT , Beddow SA , Iwasaki T , et al. Fasting hyperglycemia is not associated with increased expression of PEPCK or G6Pc in patients with type 2 diabetes.

Perry RJ , Wang Y , Cline GW , et al. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation. Miller RA , Chu Q , Xie J , Foretz M , Viollet B , Birnbaum MJ. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP.

Johanns M , Lai YC , Hsu MF , et al. AMPK antagonizes hepatic glucagon-stimulated cyclic AMP signalling via phosphorylation-induced activation of cyclic nucleotide phosphodiesterase 4B.

Nat Commun. Perry RJ , Zhang XM , Zhang D , et al. Leptin reverses diabetes by suppression of the hypothalamic-pituitary-adrenal axis. Keech DB , Utter MF. Pyruvate carboxylase: II. Krebs HA , Speake RN , Hems R. Acceleration of renal gluconeogenesis by ketone bodies and fatty acids.

Jitrapakdee S , St Maurice M , Rayment I , Cleland WW , Wallace JC , Attwood PV. Structure, mechanism and regulation of pyruvate carboxylase.

Buettner C , Patel R , Muse ED , et al. Severe impairment in liver insulin signaling fails to alter hepatic insulin action in conscious mice. Levine R , Fritz IB. The relation of insulin to liver metabolism. Fraze E , Donner CC , Swislocki ALM , Chiou YAM , Chen YDI , Reaven GM.

J Clin Endocrinol Metabol. Swislocki ALM , Chen YDI , Golay A , Chang MO , Reaven M. Insulin suppression of plasma-free fatty acid concentration in normal individuals and patients with Type 2 non-insulin-dependent diabetes.

Chen YDI , Golay A , Swislocki ALM , Reaven GM. Previs SF , Cline GW , Shulman GI. A critical evaluation of mass isotopomer distribution analysis of gluconeogenesis in vivo.

Am J Physiol. Ader M , Bergman RN. Peripheral effects of insulin dominate suppression of fasting hepatic glucose production. Rebrin K , Steil GM , Mittelman SD , Bergman RN. Causal linkage between insulin suppression of lipolysis and suppression of liver glucose output in dogs. Giacca A , Fisher SJ , Shi ZQ , Gupta R , Lickley HL , Vranic M.

Importance of peripheral insulin levels for insulin-induced suppression of glucose production in depancreatized dogs. Sindelar DK , Balcom JH , Chu CA , Neal DW , Cherrington AD. A comparison of the effects of selective increases in peripheral or portal insulin on hepatic glucose production in the conscious dog.

Puhakainen I , Koivisto VA , Yki-Järvinen H. Lipolysis and gluconeogenesis from glycerol are increased in patients with noninsulin-dependent diabetes mellitus.

Nurjhan N , Consoli A , Gerich J. Increased lipolysis and its consequences on gluconeogenesis in non-insulin-dependent diabetes mellitus. Jomain-Baum M , Hanson RW. Regulation of hepatic gluconeogenesis in the guinea pig by fatty acids and ammonia.

Sugano T , Shiota M , Tanaka T , Miyamae Y , Shimada M , Oshino N. Intracellular redox state and stimulation of gluconeogenesis by glucagon and norepinephrine in the perfused rat liver. J Biochem. Neeland IJ , Hughes C , Ayers CR , Malloy CR , Jin ES.

Effects of visceral adiposity on glycerol pathways in gluconeogenesis. Chappell J. Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC.

Boudaba, N. AMPK re-activation suppresses hepatic steatosis but its downregulation does not promote fatty liver development. EBioMedicine 28, — Candido, S. Metformin influences drug sensitivity in pancreatic cancer cells. Cao, J. Low concentrations of metformin suppress glucose production in hepatocytes through AMP-activated protein kinase AMPK.

Cokorinos, E. Activation of skeletal muscle AMPK promotes glucose disposal and glucose lowering in non-human primates and mice. Dilman, V. Metabolic immunodepression and metabolic immunotherapy: an attempt of improvement in immunologic response in breast cancer patients by correction of metabolic disturbances.

Oncology 39, 13— Inhibition of DMBA-induced carcinogenesis by phenformin in the mammary gland of rats. PubMed Abstract Google Scholar. Dowling, R. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells.

Duca, F. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats. Evans, J. Metformin and reduced risk of cancer in diabetic patients.

BMJ , — Ford, R. Metformin and salicylate synergistically activate liver AMPK, inhibit lipogenesis and improve insulin sensitivity. Foretz, M. Forslund, K. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota.

Nature , — Franciosi, M. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS One 8:e Fullerton, M. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin.

Garg, S. Diabetes and cancer: two diseases with obesity as a common risk factor. Diabetes Obes. Gui, D. Gwinn, D. AMPK phosphorylation of raptor mediates a metabolic checkpoint.

Cell 30, — Hadad, S. Evidence for biological effects of metformin in operable breast cancer: a pre-operative, window-of-opportunity, randomized trial. Breast Cancer Res. Evidence for biological effects of metformin in operable breast cancer: biomarker analysis in a pre-operative window of opportunity randomized trial.

Hardie, D. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Cell Biol. He, G. AMP-activated protein kinase induces p53 by phosphorylating MDMX and inhibiting its activity.

He, J. Metformin suppressed the proliferation of LoVo cells and induced a time-dependent metabolic and transcriptional alteration. He, L. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein. Cell , — Metformin action: concentrations matter.

Hirsch, H. Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Hosono, K. Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial.

Cancer Prev. Hou, G. Clinical pathological characteristics and prognostic analysis of 1, breast cancer patients with diabetes. Howell, J. Metformin inhibits hepatic mTORC1 signaling via dose-dependent mechanisms involving AMPK and the TSC complex.

Huai, L. Iliopoulos, D. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types.

Inoki, K. TSC2 mediates cellular energy response to control cell growth and survival. Inzucchi, S. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. Jensen, J. Diabetes Metab. Jiralerspong, S. Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer.

Jones, R. AMP-activated protein kinase induces a pdependent metabolic checkpoint. Cell 18, — Kalender, A. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner.

Kalogirou, C. Metformin-derived growth inhibition in renal cell carcinoma depends on miRmediated PTEN expression.

Karube, Y. Reduced expression of Dicer associated with poor prognosis in lung cancer patients. Cancer Sci. Kim, Y. Metformin inhibits hepatic gluconeogenesis through AMP-activated protein kinase-dependent regulation of the orphan nuclear receptor SHP.

Kordes, S. Metformin in patients with advanced pancreatic cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol.

Laplante, M. mTOR signaling in growth control and disease. Li, Z. Metformin inhibits the proliferation and metastasis of osteosarcoma cells by suppressing the phosphorylation of Akt. Lin, S. AMPK: sensing glucose as well as cellular energy status.

Liu, X. Metformin targets central carbon metabolism and reveals mitochondrial requirements in human cancers. Luft, D. Lactic acidosis in biguanide-treated diabetics: a review of cases.

Diabetologia 14, 75— Madiraju, A. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Mamtani, R. Incidence of bladder cancer in patients with type 2 diabetes treated with metformin or sulfonylureas. Diabetes Care 37, — Martello, G.

Mechanisms of Action of Metformin | IntechOpen Metformin mechanism of action between malate-aspartate Metformin mechanism of action and citric acid cycle in jechanism heart mitochondria. Inside the cell metformin inhibits Metformjn respiratory-chain complex 1, resulting Metformib reduced ATP mchanism and increased AMP. However, despite initially lower birth weight, children exposed to metformin during pregnancy had accelerated growth after birth, and were heavier by mid-childhood than those exposed to insulin during pregnancy. Effect on longevity: metformin as an elixir of life? Archived from the original on 8 June Brain Plasticity.
Molecular mechanisms of action of metformin: latest advances and therapeutic implications Impact of intermediate hyperglycemia and diabetes on immune dysfunction in tuberculosis. Metformin-associated lactate production may also take place in the large intestine, which could potentially contribute to lactic acidosis in those with risk factors. Drug Saf. In this mini-review, we discuss the latest cutting-edge research discoveries on novel molecular targets of metformin in glycemic control, cardiovascular protection, cancer intervention, anti-inflammation, antiaging, and weight control. a , Following putative transporter-mediated internalization in various immune cell subsets, metformin inhibits the mitochondrial respiratory chain complex I and can modulate cell-specific inflammatory processes by both AMP-activated protein kinase AMPK -independent and AMPK-dependent mechanisms. Gin H , Messerchmitt C , Brottier E , Aubertin J.
Mechanisms of Action of Metformin

Two studies showed that portal infusions of insulin in dogs reduced HGP; however, suppression of HGP tracked very closely with peripheral insulin levels rather than portal insulin , Further evidence for indirect regulation of gluconeogenesis came from studies reporting increased glycerol turnover and gluconeogenesis from glycerol in type 2 diabetic patients compared with healthy controls , Additionally, infusion of acetate and glycerol to maintain intrahepatic acetyl-CoA concentration and glycerol turnover respectively in awake rodents is sufficient to prevent insulin suppression of hepatic gluconeogenesis, indicating that regulation of WAT lipolysis by insulin indirectly regulates hepatic gluconeogenesis and maintains euglycemia One proposed mechanism of action for metformin is inhibition of GPD2, leading to reduced hepatic gluconeogenesis in a substrate-specific, redox-dependent manner.

This is supported by the observation that acute metformin treatment increases plasma glycerol and hepatic glycerolphosphate G3P concentrations in rodents, indicating reduced gluconeogenesis from glycerol 48 , At clinically relevant concentrations, metformin is shown to inhibit GPD2, leading to increased cytosolic redox state and decreasing gluconeogenesis from redox-dependent substrates.

Redox balance is maintained by the continuous function of 2 redox shuttles: the malate-aspartate shuttle and the α-glycerophosphate shuttle. Perturbation of this balance of reducing equivalents can directly impact gluconeogenesis from redox-dependent substrates.

This regulatory mechanism is especially pertinent in the context of obesity and T2D due to dysregulated WAT lipolysis and increased glycerol supply to the liver , , Therefore, inhibition of gluconeogenesis from glycerol may disproportionately benefit individuals with poorly controlled T2D with dysregulated WAT lipolysis.

Importantly, this substrate-specific effect that is observed with metformin treatment is not predicted by any other proposed mechanisms of metformin action 48 , Furthermore, this substrate-specific effect of metformin to inhibit gluconeogenesis would also explain why hypoglycemia is rarely observed in patients taking metformin given that other substrates eg, alanine and other amino acids are still able to contribute to gluconeogenesis.

This proposed mechanism of action will be discussed further below. This hypothesis emerged in the early s when 2 groups reported robust inhibition of complex I following metformin treatment in vitro 57 , These data are consistent with previous studies performed more than 50 years ago demonstrating that phenformin and other guanides inhibit complex I activity Complex I is the site of NADH contribution to the mitochondrial proton gradient and, given the energetic cost of gluconeogenesis, its inhibition was linked to decreased HGP.

Several mechanisms by which complex I inhibition leads to suppression of gluconeogenesis are proposed, including altered hepatic energy charge and AMPK activation 98 , , However, the physiological relevance of these mechanisms has been contested due to the supra-pharmacological millimolar concentrations typically used in these studies 48 , 50 , Additionally, these studies are challenged by conflicting data showing that metformin does not alter hepatic energy charge and does not require AMPK activation to exert its therapeutic effects in vivo 59 , In this section, we review the studies related to complex I inhibition by metformin.

Proposed mechanisms of metformin action. Complex I inhibition top panel. Inhibition of complex I is central to several proposed mechanisms of metformin action. Following complex I inhibition, AMPK is activated by increased AMP levels, leading to inhibition of CRTC2 and preventing formation of the CREB-CBP-CRTC2 complex orange boxes.

AMPK also phosphorylates and inhibits ACC1 and 2, which promotes fat oxidation and decreases lipogenesis purple boxes. Additionally, increased AMP is proposed to inhibit hepatic gluconeogenesis independently of AMPK.

High AMP prevents glucagon-stimulated production of cAMP and therefore antagonizes hepatic glucagon action pink boxes. AMP also allosterically inhibits FBP1, which directly inhibits the gluconeogenic pathway blue boxes. Increased cytosolic redox bottom panel.

Metformin inhibition of GPD2 is the only proposed mechanism that is independent of Complex I inhibition and produces substrate-selective inhibition of gluconeogenesis. Abbreviations: ACC, acetyl-CoA carboxylase; AMPK, adenosine monophosphate—activated protein kinase; CBP, CREB-binding protein; CREB, cAMP-responsive element-binding protein 1; CRTC2, CREB-regulated transcription co-activator 2; DHAP, dihydroxyacetone phosphate; FBP, fructose 1,6-bisphosphatase; FDP, fructose 1,6-diphosphate; G3P, glycerolphosphate; GLP-1, glucagon-like peptide-1; GPD, glycerolphosphate dehydrogenase; LDH, lactate dehydrogenase; OCT1, organic cation transporter 1.

Complex I inhibition at millimolar concentrations of metformin is well-established, and metformin is proposed to alter hepatic adenine nucleotide energy charge due to decreased electron transport chain activity 58 , Specifically, reduced electron transport chain activity decreases the cellular [ATP]:[ADP] and [ATP]:[AMP] ratios, which may potentially mediate the antidiabetic effects of metformin.

One study reported that increased hepatic AMP concentrations following metformin treatment allosterically inhibits adenylyl cyclase, decreasing intracellular cAMP production and antagonizing hepatic glucagon action However, we and others have not observed an effect of metformin on cAMP levels at clinically relevant concentrations of metformin 50 , 59 , Moreover, a trial of metformin in individuals with prediabetes showed that metformin did not suppress glucagon-dependent HGP, demonstrating that this mechanism for metformin action does not appear to be relevant to humans It should be noted that Miller et al propose an additional mechanism, in which AMP directly inhibits gluconeogenesis through allosteric inhibition of fructose 1,6-biphosphatase In support of this mechanism, a recent study reported that expression of a mutant fructose 1,6-bisphosphatase enzyme that is not regulated by AMP abrogated the glucose-lowering effect of metformin in vivo Thus, further investigation is needed to determine the clinical significance of this mechanism.

Whether clinically relevant concentrations of metformin alter hepatocellular adenine nucleotide levels is also unresolved.

As discussed earlier, one of the main challenges in interpreting these studies as well as other studies that have implicated complex I as the major therapeutic target of metformin is the supra-pharmacological doses of metformin utilized in many studies , 63 , , Tables 1 , 2.

Initial studies indicated that, in vitro, the K 0. Subsequent studies have observed complex I inhibition only in the presence of 2 mM to 10 mM and, similarly, altered hepatic energy charge is not observed at clinically relevant concentrations, which are in the micromolar range 58 , 59 , Furthermore, studies in lean and rat models of T2D report no change in hepatic [ATP]:[ADP] or [ATP]:[AMP] ratios following acute or chronic metformin treatment 48 , In addition to complex I inhibition, one of the most frequently invoked mechanisms of metformin action is AMPK activation due to complex I inhibition.

This occurs during times of metabolic stress, such as prolonged starvation and intense exercise , The proposed beneficial metabolic effects of AMPK activation are two-fold: transcriptional downregulation of gluconeogenic genes reduces HGP, and phosphorylation of acetyl-CoA carboxylase 1 ACC1 and ACC2, which reduces lipogenesis and promotes hepatic mitochondrial oxidation, respectively , resulting in reductions in hepatic diacylglycerol content and improved hepatic insulin sensitivity , , Although there is disagreement in the literature regarding hepatic energy charge as a mechanism of metformin action, metformin is shown to activate AMPK-Thr phosphorylation independently of changes in adenine nucleotides 48 , , However, it is unlikely that metformin directly binds to and activates AMPK because metformin has no effect on the activity of purified AMPK In the search for a new mechanism of AMPK activation, LKB1 was identified in the early s as an upstream kinase responsible for phosphorylating and activating AMPK and was implicated as a major target of metformin action , Activation of this pathway induces disassembly of the CREB-CBP-CRTC2 complex which transcriptionally regulates gluconeogenic gene expression 91 , In support of this mechanism, liver-specific LKB1 knockout mice presented with hyperglycemia, inactivation of AMPK, and transcriptional upregulation of gluconeogenesis.

However, conflicting data were reported by Foretz et al, demonstrating that LKB1 knockout hepatocytes were surprisingly responsive to metformin therapy, and liver-specific deletion of AMPK was insufficient to suppress metformin action The notable discrepancies between these studies may be due to variation in metformin dosage, route of administration intraperitoneal versus intragastric , and diet composition regular chow versus high fat.

ACC1 and ACC2 are major downstream targets of AMPK activation that are involved with the regulation of lipid metabolism. Specifically, ACC1 and ACC2 catalyze the production of malonyl-CoA, a precursor for de novo lipogenesis and a regulator of mitochondrial fat oxidation Thus, phosphorylation and inhibition of ACC1 and ACC2 by AMPK decreases hepatic lipogenesis and increases hepatic fat oxidation respectively , leading to reduced hepatic lipid accumulation and improved insulin sensitivity.

In an elegant study using ACC double knock-in DKI mice that are insensitive to AMPK inhibition, Fullerton et al showed that AMPK inhibition of ACC is necessary for the therapeutic actions of chronic metformin treatment in mice fed a high-fat diet Interestingly, the authors show that these mice were in fact responsive to acute metformin treatment.

This is consistent with a later study using the same ACC DKI mouse model, which showed no genotype differences in response to acute metformin treatment following high-fat feeding 48 , suggesting an ACC-independent mechanism. Indeed, multiple groups have shown that both pharmacological inhibition of hepatic ACC and activation of AMPK independently reverse hepatic steatosis and restore hepatic insulin sensitivity in rodents, nonhuman primates, and humans In contrast, metformin is unable to reverse hepatic steatosis or improve liver function in the absence of weight loss in nondiabetic patients Taken together, these data suggest that hepatic ACC inhibition is not a major target of metformin action in humans.

In summary, there is competing evidence both in support of and in opposition to an AMPK-dependent mechanism of action for therapeutic doses of metformin. A more recently proposed mechanism of action of metformin is increased cytosolic redox due to inhibition of hepatic GPD2 activity Fig.

In the liver, glycerol is phosphorylated to G3P and converted to DHAP by GPD2. Thus, GPD2 is necessary for glycerol entry into the gluconeogenic pathway. GPD2 is also a redox-dependent enzyme that is a key component of the α-glycerophosphate shuttle, 1 of the 2 major redox shuttles the second being the malate-aspartate shuttle.

Importantly, GPD2 inhibition reduces gluconeogenesis from redox-dependent substrates only glycerol and lactate , which differentiates this mechanism from what would be expected with complex I inhibition as well as all other proposed mechanisms for metformin action.

This substrate selectivity for metformin inhibition of hepatic gluconeogenesis has been demonstrated both in vitro and in vivo 48 , Contrary to these findings, Calza et al did not report any changes in gluconeogenesis from lactate in response to metformin in the perfused rat liver and Alshawi et al did not observe significant inhibition of GPD2 by metformin However, in contrast to these 2 studies, several other groups have independently reported an inhibitory effect of metformin and phenformin on GPD2 activity at clinically relevant μM concentrations 50 , Metformin inhibition of GPD2 activity is further supported by studies showing increased hepatocellular G3P and glycerol concentrations following metformin treatment in vitro and in vivo, which is consistent with inhibition of GPD2 activity 50 , GPD2 is widely expressed throughout the body; however, expression levels vary notably between tissues A recent report from MacDonald et al questioned the proposed GPD2-dependent mechanism of metformin action in the liver, in part due to the high level of pancreatic GPD2 expression However, this conclusion fails to consider the tissue distribution of metformin.

As discussed earlier, metformin primarily accumulates in the liver, kidney, and small intestine due to the expression profile of the OCT1, OCT3, and MATE1 transporters, which are required for metformin uptake 64 , 65 , , Indeed, metformin treatment was recently shown to alter redox balance in the kidney in addition to the liver 48 , , consistent with GPD2 inhibition in tissues in which metformin accumulates It has also been suggested that metformin inhibits the malate-aspartate shuttle; however, this mechanism is challenged by the absence of an effect of metformin on malate dehydrogenase or aspartate aminotransferase 50 , As described in the preceding sections, glycerol turnover is increased in individuals with T2D due to insulin resistance and inflammation in WAT, which in turn leads to increased contributions of glycerol to gluconeogenesis by a substrate-push mechanism , , There is also evidence to suggest that metformin can alter redox balance even when rates of the glycerophosphate shuttle do not exceed that of the malate-aspartate shuttle Data obtained from GPD2 knockout mice and patients with GPD2 mutations or deficiency have provided insight to the metabolic consequences of GPD2 inhibition 50 , Most striking is the observation that GPD2 knockout mice are protected from diet-induced hyperglycemia independent of glucose-stimulated insulin secretion Perturbation of the glycerophosphate shuttle also inhibits gluconeogenesis from glycerol, leading to impaired lipid and amino acid metabolism in mice.

The clinical relevance of these changes is demonstrated by the association between low GPD2 expression and hepatic steatosis in patients with and without NAFLD, potentially indicating reduced gluconeogenesis from glycerol In the following sections, we summarize the evidence for a direct versus indirect mechanism of inhibition, in addition to the redox-dependency of this mechanism.

Metformin is shown to decrease hepatic gluconeogenesis from redox-dependent substrates through inhibition of GPD2 activity; however, it is unclear whether metformin directly or indirectly inhibits GPD2 48 , Importantly, metformin inhibition of GPD2 exhibits noncompetitive kinetics with a K i of ~50µM, which is in stark contrast with the millimolar concentrations required to inhibit complex I activity.

Supporting a direct interaction between metformin and GPD2, independent studies using intact mitochondria, mitochondrial lysates, and isolated enzyme assays with immunoprecipitated GPD2, biguanides are shown to inhibit GPD2 activity in vitro 50 , , , although other groups have not observed an inhibitory effect Several alternative hypotheses in support of an indirect mechanism of GPD2 inhibition have also been postulated.

It is likely that metformin has many additional effects and may in fact alter the activity of several complexes of the electron transport chain, which in turn indirectly leads to inhibition of GPD2. Metformin and other guanides have consistently been shown to bind metal ions, such as copper and iron, possibly by acting as a Schiff base, which is consistent with reports that metformin interacts with heme or cytochrome c directly Indeed, downstream inhibition of the electron transport chain is shown to backlog the entire electron transport chain, providing a potential link between metformin interaction with downstream complexes and indirect inhibition of GPD2 , GPD2 is also calcium-dependent and is therefore sensitive to changes in local calcium concentrations; however, there is little evidence to support a role for metformin in calcium homeostasis Further investigation is necessary to determine whether metformin and other guanides interact with copper and iron contained in the electron transport chain, which in turn results in inhibition of GPD2 by an indirect mechanism leading to an increase in the cytosolic redox state.

Metformin-metal interactions might also explain the pleotropic effects of metformin on mitochondrial function and other metabolic processes. The observation that metformin selectively inhibits gluconeogenesis from redox-dependent substrates lactate and glycerol , but not redox-independent substrates pyruvate, DHAP, alanine strongly indicates a redox-dependent mechanism of action 37 , , Several groups have shown that biguanide treatment increases the cytosolic redox state in a variety of tissues, consistent with inhibition of the glycerophosphate shuttle 48 , , , , , , , Metformin-induced increases in the cytosolic redox state will reduce lactate conversion to pyruvate due to the dependency of lactate dehydrogenase activity on the cytosolic redox state and it will reduce glycerol conversion to glucose due to inhibition of GPD2 activity.

Isotopically labeled tracer studies have independently confirmed that metformin reduces gluconeogenesis from lactate and glycerol, while gluconeogenesis from alanine remains unaltered 37 , 48 , Importantly this substrate-selective inhibition of gluconeogenesis cannot be explained by any of the previously described proposed mechanisms of metformin action, including complex I inhibition, AMPK activation, fructose 1,6-bisphosphatase inhibition, or CREB inhibition.

In comparison with other pharmacologic interventions for T2D, metformin treatment rarely causes hypoglycemia Yet, this remains an area of active investigation.

Here, we have highlighted several characteristics of metformin action that are consistent with a redox-dependent mechanism of action on hepatic gluconeogenesis at therapeutically relevant μM concentrations.

Furthermore, we described key features of metformin treatment that are inconsistent with clinically meaningful complex I inhibition, including the millimolar concentrations required to observe complex I inhibition, and the substrate selectivity of metformin inhibition of gluconeogenesis.

Many of the postulated mechanisms of metformin action, including AMPK activation and altered energy charge, are dependent on significant complex I inhibition. Therefore, although these pathways may play a role in the pleiotropic effects of metformin, they are likely dispensable for the glucose-lowering effect of metformin in patients with poorly controlled T2D.

In recent years, the utility of metformin has been expanded beyond the first-line treatment for T2D, and it has the potential to enter the realms of aging, cancer, and cardiovascular disease.

Thus, the quest to identify a clear mechanism of metformin action is pertinent to the development of new therapeutic strategies and alleviating these chronic illnesses. Financial Support: This work was supported by grants from the United States Public Health Service R01 DK, R01 DK, R01 DK, R01 DK, RC2 DK, P30 DK, and GM Disclosure Summary: G.

serves on the scientific advisory boards for Merck, AstraZeneca, iMBP, and Janssen Research and Development and receives investigator-initiated support from Gilead Sciences, Merck, and AstraZeneca. He is also a Scientific Co-Founder of TLC.

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study. Diabetes Care.

King P , Peacock I , Donnelly R. The UK prospective diabetes study UKPDS : clinical and therapeutic implications for type 2 diabetes. Br J Clin Pharmacol. Google Scholar. DeFronzo RA , Goodman AM. Efficacy of metformin in patients with non-insulin-dependent diabetes mellitus.

N Engl J Med. Hill J. The Vegetable System. London : R. Google Preview. Bailey CJ. Metformin: historical overview. Watanabe CK. Studies in the metabolism changes induced by administration of guanidine bases: I.

influence of injected guanidine hydrochloride upon blood sugar content. J Biol Chem. Werner EA , Bell J. J Chem Soc Trans. Sterne J. Blood sugar-lowering effect of 1,1-dimethylbiguanide. Knaani Z , Klajman A. Our experiences with N,N-dimethyl-guanyl-guanidine La in the treatment of diabetic patients.

Bischoff F , Long ML. Guanidine structure and hypoglycemia. J Pharmacol Exp Ther. McKendry JB , Kuwayti K , Rado PP.

Clinical experience with DBI phenformin in the management of diabetes. Can Med Assoc J. Ungar G , Freedman L , Shapiro SL.

Pharmacological studies of a new oral hypoglycemic drug. Proc Soc Exp Biol Med. Wien Med Wochenschr. Hermann LS. Metformin: a review of its pharmacological properties and therapeutic use. Diabete Metab. Schäfer G. A review of history, pharmacodynamics and therapy. Reaven GM. Role of insulin resistance in human disease.

Bailey CJ , Puah JA. Effect of metformin on glucose metabolism in mouse soleus muscle. Jackson RA , Hawa MI , Jaspan JB , et al. Mechanism of metformin action in non-insulin-dependent diabetes. Pagano G , Tagliaferro V , Carta Q , et al. Metformin reduces insulin requirement in Type 1 insulin-dependent diabetes.

Pedersen O , Nielsen O , Bak J , Richelsen B , Beck-Nielsen H , Sørensen N. The effects of metformin on adipocyte insulin action and metabolic control in obese subjects with type 2 diabetes. Diabet Med. Gin H , Messerchmitt C , Brottier E , Aubertin J. Metformin improved insulin resistance in type I, insulin-dependent, diabetic patients.

World Health Organization. WHO model list of essential medicines. World Health Organization ; Accessed April 30, Evans JM , Donnelly LA , Emslie-Smith AM , Alessi DR , Morris AD. Metformin and reduced risk of cancer in diabetic patients. Zakikhani M , Dowling R , Fantus IG , Sonenberg N , Pollak M.

Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res. Barzilai N , Crandall JP , Kritchevsky SB , Espeland MA. Metformin as a Tool to Target Aging. Cell Metab. Justice JN , Ferrucci L , Newman AB , et al. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup.

Bharath LP , Agrawal M , McCambridge G , et al. Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation.

Cell Metab ; 32 : 44 - Proc Natl Acad Sci U S A. Petrie JR , Chaturvedi N , Ford I , et al. Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes REMOVAL : a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol.

Maruthur NM , Tseng E , Hutfless S , et al. Diabetes medications as monotherapy or metformin-based combination therapy for type 2 diabetes: a systematic review and meta-analysis. Ann Intern Med. Grant PJ. The effects of metformin on cardiovascular risk factors. Diabetes Metab Rev.

Juhan-Vague I , Vague P , Alessi MC , et al. Relationships between plasma insulin triglyceride, body mass index, and plasminogen activator inhibitor 1.

Rains SG , Wilson GA , Richmond W , Elkeles RS. The reduction of low density lipoprotein cholesterol by metformin is maintained with long-term therapy.

J R Soc Med. McCreight LJ , Mari A , Coppin L , Jackson N , Umpleby AM , Pearson ER. Metformin increases fasting glucose clearance and endogenous glucose production in non-diabetic individuals. Galuska D , Nolte LA , Zierath JR , Wallberg-Henriksson H. Effect of metformin on insulin-stimulated glucose transport in isolated skeletal muscle obtained from patients with NIDDM.

Inzucchi SE , Maggs DG , Spollett GR , et al. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. Stumvoll M , Nurjhan N , Perriello G , Dailey G , Gerich JE. Metabolic effects of metformin in non-insulin-dependent diabetes mellitus.

Samuel Varman T , Shulman Gerald I. Mechanisms for insulin resistance: common threads and missing links. Magnusson I , Rothman DL , Katz LD , Shulman RG , Shulman GI. Increased rate of gluconeogenesis in type II diabetes mellitus. J Clin Invest. Nosadini R , Avogaro A , Trevisan R , et al. Effect of metformin on insulin-stimulated glucose turnover and insulin binding to receptors in type II diabetes.

Johnson AB , Webster JM , Sum CF , et al. The impact of metformin therapy on hepatic glucose production and skeletal muscle glycogen synthase activity in overweight type II diabetic patients. Hundal RS , Krssak M , Dufour S , et al. Mechanism by which metformin reduces glucose production in type 2 diabetes.

Widén EI , Eriksson JG , Groop LC. Metformin normalizes nonoxidative glucose metabolism in insulin-resistant normoglycemic first-degree relatives of patients with NIDDM.

Forslund K , Hildebrand F , Nielsen T , et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Wu H , Esteve E , Tremaroli V , et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug.

Nat Med. Graham GG , Punt J , Arora M , et al. Clinical pharmacokinetics of metformin. Clin Pharmacokinet. Timmins P , Donahue S , Meeker J , Marathe P. Steady-state pharmacokinetics of a novel extended-release metformin formulation.

Madiraju AK , Qiu Y , Perry RJ , et al. Metformin inhibits gluconeogenesis via a redox-dependent mechanism in vivo. Kajbaf F , De Broe ME , Lalau JD. Therapeutic concentrations of metformin: a systematic review. Madiraju AK , Erion DM , Rahimi Y , et al.

Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Frid A , Sterner GN , Löndahl M , et al. Novel assay of metformin levels in patients with type 2 diabetes and varying levels of renal function: clinical recommendations.

Wang Y , Nasiri AR , Damsky WE , et al. Uncoupling hepatic oxidative phosphorylation reduces tumor growth in two murine models of colon cancer. Cell Rep. Chandel NS , Avizonis D , Reczek CR , et al. Are metformin doses used in murine cancer models clinically relevant?

Wilcock C , Bailey CJ. Accumulation of metformin by tissues of the normal and diabetic mouse. He L , Wondisford FE. Metformin action: concentrations matter. Gormsen LC , Sundelin EI , Jensen JB , et al. In vivo imaging of human 11 C-Metformin in peripheral organs: dosimetry, biodistribution, and kinetic analyses.

J Nucl Med. Owen MR , Doran E , Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. Metformin might influence tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction of energetic stress; however, these effects require further investigation.

Here, we discuss the updated understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries of metformin targets for the treatment of diabetes mellitus and cancer.

Abstract Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the mechanistic aspects of metformin action remained ill-defined. Publication types Research Support, Non-U. Show pagesource. Old revisions. Recent Changes.

Metformin Trade Names: Glucophage, Glucophage XR ®. decreases the liver's production of glucose via activation of AMP-activated protein kinase AMPK see Figure. at very high doses it may increase the removal of glucose from muscle, the liver, and other body tissues where it is stored.

a FIRST LINE drug in the treatment of type II diabetes except when contraindicated - see below. it has been shown to reduce the risk of cardiovascular events and macrovascular disease Kooy et al, ; Wexler, not as an FDA approved indication in obese type 1 diabetics as an adjunct to insulin not as monotherapy to reduce weight and to better control hemoglobin A1C levels.

IMPORTANT Contraindications :. chronic cardiopulmonary dysfunction congestive heart failure, emphysema - conditions that predispose patients to tissue anoxia, which increases the risk of lactic acidosis.

Previous guidelines had contraindications based upon serum creatinine levels vs estimated GFR:. liver disease because of increased risk of lactic acidosis. There isn't a fixed dosage regimen for metformin. It can be given in divided doses with meals, or as an extended-release formulation Glucophage XR ® that is generally given once daily with the evening meal.

Rare : dose-related incidence of lactic acidosis. Vitamin B12 deficiency. In rare patients, Vit B12 deficiency may result in peripheral neuropathy and megaloblastic anemia. Taking a daily multivitamin may protect against Vit B12 deficiency McCulloch, Most patients with type 2 diabetes, as well as those with prediabetes are overweight or obese.

Metformin has been found to produce a more significant degree of weight loss in patients with polycystic ovary syndrome PCOS , a complex endocrine disorder with symptoms including diabetes, obesity, infertility and heart disease Li et al, metformin can result in lactic acidosis due to its effect to block gluconeogenesis, which can impair the hepatic metabolism of lactic acid.

Anderson K et al : Is metformin effective for reducing weight in obese or overweight adolescents?. DOI:

Metformin mechanism of action

Author: Vile

0 thoughts on “Metformin mechanism of action

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com