Metformin improves healthspan and lifespan in mice

Alejandro Martin-Montalvo, Evi M Mercken, Sarah J Mitchell, Hector H Palacios, Patricia L Mote, Morten Scheibye-Knudsen, Ana P Gomes, Theresa M Ward, Robin K Minor, Marie-José Blouin, Matthias Schwab, Michael Pollak, Yongqing Zhang, Yinbing Yu, Kevin G Becker, Vilhelm A Bohr, Donald K Ingram, David A Sinclair, Norman S Wolf, Stephen R Spindler, Michel Bernier, Rafael de Cabo, Alejandro Martin-Montalvo, Evi M Mercken, Sarah J Mitchell, Hector H Palacios, Patricia L Mote, Morten Scheibye-Knudsen, Ana P Gomes, Theresa M Ward, Robin K Minor, Marie-José Blouin, Matthias Schwab, Michael Pollak, Yongqing Zhang, Yinbing Yu, Kevin G Becker, Vilhelm A Bohr, Donald K Ingram, David A Sinclair, Norman S Wolf, Stephen R Spindler, Michel Bernier, Rafael de Cabo

Abstract

Metformin is a drug commonly prescribed to treat patients with type 2 diabetes. Here we show that long-term treatment with metformin (0.1% w/w in diet) starting at middle age extends healthspan and lifespan in male mice, while a higher dose (1% w/w) was toxic. Treatment with metformin mimics some of the benefits of calorie restriction, such as improved physical performance, increased insulin sensitivity, and reduced low-density lipoprotein and cholesterol levels without a decrease in caloric intake. At a molecular level, metformin increases AMP-activated protein kinase activity and increases antioxidant protection, resulting in reductions in both oxidative damage accumulation and chronic inflammation. Our results indicate that these actions may contribute to the beneficial effects of metformin on healthspan and lifespan. These findings are in agreement with current epidemiological data and raise the possibility of metformin-based interventions to promote healthy aging.

Figures

Figure 1. Metformin increases survival and improves…
Figure 1. Metformin increases survival and improves physical performance
(a, b) Kaplan–Meier survival curve for mice treated either with 0.1% or 1% metformin. n = 64 for metformin 0.1% group and n = 83 for their untreated counterparts; n = 90 for metformin 1% group and n = 88 for their untreated counterparts. The arrows at 54 weeks indicate the age at which metformin treatment was initiated. (c) Body weights. (d) Food consumption. (e, f) In vivo metabolic response to 0.1% metformin treatment. n = 9 per group. (e) Energy expenditure. (f) Respiratory exchange ratio. (g) Time to fall from an accelerating rotarod. n = 16 per group. (h) Distance ran on treadmill performance. n = 9 per group. (i) Average speed of animals in the open-field test. n = 15–16 per group. (j) Metformin treatment delayed the onset of age-related cataracts. n = 93–124 eyes per group. (k) Plasma levels of glucose after oral glucose load (OGTT). n = 8 per group. (l) Area under OGTT curve. (m) Plasma levels of glucose after intraperitoneal insulin injection (ITT). n = 9 per group. (n) Area under ITT curve. Metformin, Met. Unless otherwise stated n = all live animals in the study. Data are represented as the mean ± SEM. * p < 0.05 compared to standard diet (SD)-fed mice (t-test two tailed).
Figure 2. Metformin shifts expression patterns of…
Figure 2. Metformin shifts expression patterns of mice towards those on calorie restriction
(a) Principal component analysis (PCA) was performed on differentially expressed genes from the liver and muscle tissue of mice maintained on SD and 0.1% metformin. Each data point corresponds to the PCA analysis of each subject. (b) Gene expression profile comparing genes significantly up- (red) and down-regulated (blue) by either calorie restriction (CR) or metformin compared to SD mice (z-ratio). The percentage of significant gene expression changes shifted in the same direction in CR and metformin treatments compared to SD mice is presented in brackets. (c) Comparison of gene sets significantly altered by CR and metformin treatment compared to SD expression (z-score); up- (red) and down-regulated (blue) gene sets. The percentage of significant gene sets changes shifted in the same direction in CR and metformin treatments compared to SD mice is presented in brackets. (d) Effect of metformin on mitochondria (Mito), glycolysis, lipid metabolism (Lipid Met) and stress response (Stress Resp) related gene sets. The list of all the significantly modified gene sets can be found in Supplementary table S6. Metformin, Met; Calorie restriction, CR.
Figure 3. Metformin activates AMPK without altering…
Figure 3. Metformin activates AMPK without altering in vivo electron transport chain activities
(a) Activation of AMPK by metformin in MEFs. AMPK and acetyl-coA carboxylase (ACC) phosphorylation by metformin. n = 3 per group. (b) Activation of AMPK and ACC phosphorylation in the liver of 0.1% metformin-treated mice. n = 4–6 per group. (c) Oxygen consumption in MEFs treated with 1 mM metformin. n = 3 per group. (d, e), Mitochondrial content in MEFs treated with metformin was determined by tetramethyl rhodamine methyl ester (TMRM) (d) and MitoTracker green (e) staining, M.F.I., mean fluorescence intensity. n = 3 per group. (f) Mitochondrial DNA content analyzed by quantitative PCR in the liver. n = 5–8 per group. (g) Mitochondrial protein levels in MEFs treated with metformin. n = 3 per group. (h) Mitochondrial protein levels in the liver from 0.1% metformin-treated mice. n = 4–6 per group. (i, j) Effect of metformin on mitochondrial enzymatic activities. (i) MEFs treated with 1 mM metformin (n = 3 per group) and (j) Liver lysates from 0.1% metformin-treated mice (n = 5–6 per group). Metformin, Met: Calorie restriction, CR; Ut, untreated. Data are represented as the mean ± SEM. * p < 0.05 versus untreated controls or standard diet (SD)-fed mice (t-test two tailed).
Figure 4. Metformin enhances antioxidant defenses and…
Figure 4. Metformin enhances antioxidant defenses and inhibits inflammation
(a) Rate of electrons derived to superoxide generation in mitochondrial complexes I and II to III in the liver of 0.1% metformin-treated mice. n = 5–6 per group. (b) Oxidative damage in proteins determined by lysine-4-hydroxinonenal levels in the liver of 0.1% metformin-treated mice. (n = 4–6 per group) (c) Oxidative damage in lipids determined by 8-iso-PGF2α levels in the liver of 0.1% metformin-treated mice. (n = 4–6 per group) (d) Nrf2-ARE assay determining Nrf2-ARE-dependent expression in metformin-treated HepG2 cells. tBHQ was added as positive control for NRF2-ARE induction. (n = 3 per group) (e) Antioxidant and stress response protein levels in the liver of 0.1% metformin-treated mice. (n = 4–6 per group) (f) Activation of pro-inflammatory signaling pathways in the liver of 0.1% metformin-treated mice. (n = 4–6 per group) (g) Expression of multiple inflammatory-related genes in the liver of 0.1% metformin-treated mice. (n = 5 per group) Metformin, Met; Calorie restriction, CR. Data are represented as the mean ± SEM. * p < 0.05 versus standard diet (SD)-fed mice (t-test two tailed).

References

    1. He L, et al. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein. Cell. 2009;137:635–646.
    1. Correia S, et al. Mechanisms of action of metformin in type 2 diabetes and associated complications: an overview. Mini Rev Med Chem. 2008;8:1343–1354.
    1. Pollak MN. Investigating Metformin for Cancer Prevention and Treatment: The End of the Beginning. Cancer Discov. 2012;9:778–790.
    1. Libby G, et al. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–1625.
    1. Giovannucci E, et al. Diabetes and cancer: a consensus report. Diabetes care. 2010;33:1674–1685.
    1. Harrison DE, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395.
    1. Baur JA, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337–342.
    1. Martin-Montalvo A, Villalba JM, Navas P, de Cabo R. NRF2, cancer and calorie restriction. Oncogene. 2011;30:505–520.
    1. Anisimov VN, et al. If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging. 2011;3:148–157.
    1. Onken B, Driscoll M. Metformin induces a dietary restriction-like state and the oxidative stress response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. PloS one. 2010;5:e8758.
    1. Smith DL, Jr, et al. Metformin supplementation and life span in Fischer-344 rats. J. Gerontol. A Biol. Sci. Med. Sci. 2010;65:468–474.
    1. Anisimov VN, et al. Gender differences in metformin effect on aging, life span and spontaneous tumorigenesis in 129/Sv mice. Aging (Albany NY) 2010;2:945–958.
    1. Slack C, Foley A, Partridge L. Activation of AMPK by the putative dietary restriction mimetic metformin is insufficient to extend lifespan in Drosophila. PLoS ONE. 2012;7:e47699.
    1. Dhahbi JM, Mote PL, Fahy GM, Spindler SR. Identification of potential caloric restriction mimetics by microarray profiling. Physiol. Genomics. 2005;23:343–350.
    1. Spindler SR. Use of microarray biomarkers to identify longevity therapeutics. Aging Cell. 2006;5:39–50.
    1. Zhou G, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 2001;108:1167–1174.
    1. Larsson O, et al. Distinct perturbation of the translatome by the antidiabetic drug metformin. Proc. Natl. Acad. Sci. U.S.A. 2012;109:8977–8982.
    1. Saeedi R, et al. Metabolic actions of metformin in the heart can occur by AMPK-independent mechanisms. Am. J. Physiol. 2008;294:H2497–H2506.
    1. Ben Sahra I, et al. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 2011;71:4366–4372.
    1. Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 2012;13:251–262.
    1. Brunmair B, et al. Thiazolidinediones, like metformin, inhibit respiratory complex I: a common mechanism contributing to their antidiabetic actions? Diabetes. 2004;53:1052–1059.
    1. El-Mir MY, et al. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J. Biol. Chem. 2000;275:223–238.
    1. Martin-Montalvo A, de Cabo R. Mitochondrial metabolic reprogramming induced by calorie restriction. Antioxid. Redox Signal. 2012
    1. Testa CM, Sherer TB, Greenamyre JT. Rotenone induces oxidative stress and dopaminergic neuron damage in organotypic substantia nigra cultures. Brain Res. 2005;134:109–118.
    1. Hariharakrishnan J, Satpute RM, Prasad GB, Bhattacharya R. Oxidative stress mediated cytotoxicity of cyanide in LLC-MK2 cells and its attenuation by alpha-ketoglutarate and N-acetyl cysteine. Toxicol. Lett. 2009;185:132–141.
    1. Algire C, et al. Metformin reduces endogenous reactive oxygen species and associated DNA damage. Cancer Prev Res (Phila) 2012;5:536–543.
    1. Halicka HD, et al. Genome protective effect of metformin as revealed by reduced level of constitutive DNA damage signaling. Aging. 2011;3:1028–1038.
    1. Hou X, et al. Metformin reduces intracellular reactive oxygen species levels by upregulating expression of the antioxidant thioredoxin via the AMPK-FOXO3 pathway. Biochem. Biophys. Res. Commun. 2010;396:199–205.
    1. Pierotti MA, et al. Targeting metabolism for cancer treatment and prevention: metformin, an old drug with multi-faceted effects. Oncogene. 2013;32:1475–1487.
    1. Holst H, Eldrup E, Guldstad NH, Bülow HH, Christensen HR. Metformin associated with lactic acidosis in treatment of type 2 diabetes. Ugeskrift for laeger. 2012;174:1598–1602.
    1. Alley DE, et al. Changes in weight at the end of life: characterizing weight loss by time to death in a cohort study of older men. Am. J. Epidemiol. 2010;172:558–565.
    1. Pearson KJ, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell metab. 2008;8:157–168.
    1. Sheldon WG, Bucci TJ, Hart RW, Turturro A. Age-related neoplasia in a lifetime study of ad libitum-fed and food-restricted B6C3F1 mice. Toxicol Pathol. 1995;23:458–476.
    1. Pyra KA, Saha DC, Reimer RA. Prebiotic fiber increases hepatic acetyl CoA carboxylase phosphorylation and suppresses glucose-dependent insulinotropic polypeptide secretion more effectively when used with metformin in obese rats. J. Nutr. 2012;142:213–220.
    1. Collier CA, Bruce CR, Smith AC, Lopaschuk G, Dyck DJ. Metformin counters the insulin-induced suppression of fatty acid oxidation and stimulation of triacylglycerol storage in rodent skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2006;291:E182–E189.
    1. Nerstedt A, et al. AMP-activated protein kinase inhibits IL-6-stimulated inflammatory response in human liver cells by suppressing phosphorylation of signal transducer and activator of transcription 3 (STAT3) Diabetologia. 2010;53:2406–2416.
    1. Radziuk J, Bailey CJ, Wiernsperger NF, Yudkin JS. Metformin and its liver targets in the treatment of type 2 diabetes. Curr. Drug Targets Immune Endocr. Metabol. Disord. 2003;3:151–169.
    1. Kim YD, et al. Metformin inhibits hepatic gluconeogenesis through AMP-activated protein kinase-dependent regulation of the orphan nuclear receptor SHP. Diabetes. 2008;57:306–314.
    1. Gonzalez-Barroso MM, et al. Fatty acids revert the inhibition of respiration caused by the antidiabetic drug metformin to facilitate their mitochondrial beta-oxidation. Biochimica et biophysica acta. 2012;1817:1768–1775.
    1. Vives M, et al. Metformin-associated lactic acidosis: incidence, diagnosis, prognostic factors and treatment. Rev. Esp. Anestesiol. Reanim. 2012;59:276–279.
    1. Larsen S, et al. Metformin-treated patients with type 2 diabetes have normal mitochondrial complex I respiration. Diabetologia. 2012;55:443–449.
    1. Kane DA, et al. Metformin selectively attenuates mitochondrial H2O2 emission without affecting respiratory capacity in skeletal muscle of obese rats. Free Radic. Biol. Med. 2010;49:1082–1087.
    1. Sum CF, et al. The effect of intravenous metformin on glucose metabolism during hyperglycaemia in type 2 diabetes. Diabet. Med. 1992;9:61–65.
    1. Szabo G, Csak T. Inflammasomes in liver diseases. J. Hepatol. 2012;57:642–654.
    1. Brasier AR. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc. Res. 2010;86:211–218.
    1. De Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett. 2008;582:97–105.
    1. Cheadle C, Vawter MP, Freed WJ, Becker KG. Analysis of microarray data using Z score transformation. J. Mol. Diagn. 2003;5:73–81.
    1. Lee C, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci. Transl. Med. 2012;4:124–127.
    1. Kim SY, Volsky DJ. PAGE: parametric analysis of gene set enrichment. BMC bioinformatics. 2005;6:144.
    1. Padilla S, et al. Demethoxy-Q, an intermediate of coenzyme Q biosynthesis, fails to support respiration in Saccharomyces cerevisiae and lacks antioxidant activity. J. Biol. Chem. 2004;279:25995–26004.
    1. Bernier M, et al. Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 protein. J. Biol. Chem. 2011;286:19270–19279.

Source: PubMed

3
Tilaa