Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase

Abstract

Metformin is considered to be one of the most effective therapeutics for treating type 2 diabetes because it specifically reduces hepatic gluconeogenesis without increasing insulin secretion, inducing weight gain or posing a risk of hypoglycaemia1,2. For over half a century, this agent has been prescribed to patients with type 2 diabetes worldwide, yet the underlying mechanism by which metformin inhibits hepatic gluconeogenesis remains unknown. Here we show that metformin non-competitively inhibits the redox shuttle enzyme mitochondrial glycerophosphate dehydrogenase, resulting in an altered hepatocellular redox state, reduced conversion of lactate and glycerol to glucose, and decreased hepatic gluconeogenesis. Acute and chronic low-dose metformin treatment effectively reduced endogenous glucose production, while increasing cytosolic redox and decreasing mitochondrial redox states. Antisense oligonucleotide knockdown of hepatic mitochondrial glycerophosphate dehydrogenase in rats resulted in a phenotype akin to chronic metformin treatment, and abrogated metformin-mediated increases in cytosolic redox state, decreases in plasma glucose concentrations, and inhibition of endogenous glucose production. These findings were replicated in whole-body mitochondrial glycerophosphate dehydrogenase knockout mice. These results have significant implications for understanding the mechanism of metformin’s blood glucose lowering effects and provide a new therapeutic target for type 2 diabetes.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Effects of acute metformin (50 mg kg−1, intravenously) treatment in Sprague-Dawley rats on plasma glucose, EGP and hepatic redox state.
Figure 2: Effects of chronic metformin (50 mg kg−1, intraperitoneally per day × 30 days) treatment in Sprague-Dawley rats on plasma glucose, EGP and hepatic redox state.
Figure 3: Guanides/biguanides non-competitively inhibit rat and human mGPD activity.
Figure 4: Effects of mGPD and cGPD ASO treatment in Sprague-Dawley rats on metformin action.

References

  1. Hundal, R. S. et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes 49, 2063–2069 (2000)

    Article  CAS  Google Scholar 

  2. Inzucchi, S. et al. Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N. Engl. J. Med. 338, 867–872 (1998)

    Article  CAS  Google Scholar 

  3. El-Mir, M.-Y. et al. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J. Biol. Chem. 275, 223–228 (2000)

    Article  CAS  Google Scholar 

  4. Owen, M. R., Doran, E. & Halestrap, A. P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 348, 607–614 (2000)

    Article  CAS  Google Scholar 

  5. Zhou, G. et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174 (2001)

    Article  CAS  Google Scholar 

  6. Shaw, R. J. et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310, 1642–1646 (2005)

    Article  ADS  CAS  Google Scholar 

  7. He, L. et al. Metformin and insulin suppress hepatic gluconeogenesis through phosphorylation of CREB binding protein. Cell 137, 635–646 (2009)

    Article  CAS  Google Scholar 

  8. Cool, B. et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 3, 403–416 (2006)

    Article  CAS  Google Scholar 

  9. Savage, D. B. et al. Reversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2. J. Clin. Invest. 116, 817–824 (2006)

    Article  CAS  Google Scholar 

  10. Fullerton, M. D. et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nature Med. 19, 1649–1654 (2013)

    Article  CAS  Google Scholar 

  11. Foretz, M. et al. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J. Clin. Invest. 120, 2355–2369 (2010)

    Article  CAS  Google Scholar 

  12. Miller, R. A. et al. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature 494, 256–260 (2013)

    Article  ADS  CAS  Google Scholar 

  13. Hawley, S. A., Gadalla, A. E., Olsen, G. S. & Hardie, D. G. The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism. Diabetes 51, 2420–2425 (2002)

    Article  CAS  Google Scholar 

  14. Yang, L. et al. Metabolomic and mass isotopomer analysis of liver gluconeogenesis and citric acid cycle. I. Interrelation between gluconeogenesis and cataplerosis; formation of methoxamates from aminooxyacetate and ketoacids. J. Biol. Chem. 283, 21978–21987 (2008)

    Article  CAS  Google Scholar 

  15. Krebs, H. A. & Gascoyne, T. The redox state of the nicotinamide-adenine dinucleotides in rat liver homogenates. Biochem. J. 108, 513–520 (1968)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Williamson, D. H., Lund, P. & Krebs, H. A. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J. 103, 514–527 (1967)

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bremer, J. & Davis, E. J. Studies on the active transfer of reducing equivalents into mitochondria via the malate-aspartate shuttle. Biochim. Biophys. Acta 376, 387–397 (1975)

    Article  CAS  Google Scholar 

  18. Cederbaum, A. I., Lieber, C. S., Beattie, D. S. & Rubin, E. Characterization of shuttle mechanisms for the transport of reducing equivalents into mitochondria. Arch. Biochem. Biophys. 158, 763–781 (1973)

    Article  CAS  Google Scholar 

  19. Garrib A & McMurray, W. C. Purification and characterization of glycerol-3-phosphate dehydrogenase (flavin-linked) from rat liver mitochondria. J. Biol. Chem. 261, 8042–8048 (1986)

    PubMed  Google Scholar 

  20. Cole, E. S., Lepp, C. A., Holohan, P. D. & Fondy, T. P. Isolation and characterization of flavin-linked glycerol-3-phosphate dehydrogenase from rabbit skeletal muscle mitochondria and comparison with the enzyme from rabbit brain. J. Biol. Chem. 253, 7952–7959 (1978)

    CAS  PubMed  Google Scholar 

  21. White, H. B., III & Kaplan, N. O. Purification and properties of two types of diphosphopyridine nucleotide-linked glycerol 3-phosphate dehydrogenases from chicken breast muscle and chicken liver. J. Biol. Chem. 244, 6031–6039 (1969)

    CAS  PubMed  Google Scholar 

  22. Sistare, F. D. & Haynes R. C. Jr The interaction between the cytosolic pyridine nucleotide redox potential and gluconeogenesis from lactate/pyruvate in isolated rat hepatocytes. J. Biol. Chem. 260, 12748–12753 (1985)

    CAS  PubMed  Google Scholar 

  23. Sugano, T. et al. Intracellular redox state and stimulation of gluconeogenesis by glucagon and norepinephrine in the perfused rat liver. J. Biochem. 87, 153–166 (1980)

    Article  CAS  Google Scholar 

  24. MacDonald, M. J. & Marshall, L. K. Mouse lacking NAD+-linked glycerol phosphate dehydrogenase has normal pancreatic beta cell function but abnormal metabolite pattern in skeletal muscle. Arch. Biochem. Biophys. 384, 143–153 (2000)

    Article  CAS  Google Scholar 

  25. Prochazka, M., Kozak, U. C. & Kozak, L. P. A glycerol-3-phosphate dehydrogenase null mutant in BALB/cHeA mice. J. Biol. Chem. 264, 4679–4683 (1989)

    CAS  PubMed  Google Scholar 

  26. Harding, J. W., Jr, Pyeritz, E. A., Copeland, E. S. & White, H. B., III Role of glycerol 3-phosphate dehydrogenase in glyceride metabolism. Effect of diet on enzyme activities in chicken liver. Biochem. J. 146, 223–229 (1975)

    Article  CAS  Google Scholar 

  27. Harding, J. W., Jr, Pyeritz, E. A., Morris, H. P. & White, H. B., III Proportional activities of glycerol kinase and glycerol 3-phosphate dehydrogenase in rat hepatomas. Biochem. J. 148, 545–550 (1975)

    Article  CAS  Google Scholar 

  28. Brown, L. J. et al. Normal thyroid thermogenesis but reduced viability and adiposity in mice lacking the mitochondrial glycerol phosphate dehydrogenase. J. Biol. Chem. 277, 32892–32898 (2002)

    Article  CAS  Google Scholar 

  29. Barberà, A. et al. A high carbohydrate diet does not induce hyperglycaemia in a mitochondrial glycerol-3-phosphate dehydrogenase-deficient mouse. Diabetologia 46, 1394–1401 (2003)

    Article  Google Scholar 

  30. Colussi, T. et al. Structure of α-glycerophosphate oxidase from Streptococcus sp.: a template for the mitochondrial α-glycerophosphate dehydrogenase. Biochemistry 47, 965–977 (2008)

    Article  CAS  Google Scholar 

  31. Faupel, R. P., Seitz, H. J., Tarnowski W, Thiemann V & Weiss C The problem of tissue sampling from experimental animals with respect to freezing technique, anoxia, stress and narcosis. A new method for sampling rat liver tissue and the physiological values of glycolytic intermediates and related compounds. Arch. Biochem. Biophys. 148, 509–522 (1972)

    Article  CAS  Google Scholar 

  32. Ayala, J. E. et al. NIH Mouse Metabolic Phenotyping Center Consortium. Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice. Dis. Model. Mech. 3, 525–534 (2010)

    Article  CAS  Google Scholar 

  33. Carmignani, M. et al. Novel hypotensive agents from Verbesina caracasana. 8. Synthesis and pharmacology of (3,4-dimethoxycinnamoyl)-N1-agmatine and synthetic analogues. J. Med. Chem. 44, 2950–2958 (2001)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Dong, M. Kahn, Y. Kosover and J. J. Hsiao for their technical support, and S. Singh and D. Gregg for discussions. This publication was supported by grants from the National Institutes of Health: R24 DK-085638, R01 DK-40936, P30 DK-45735, P30 DK-034989, U24 DK-059635, R01 DK-28348, K01 DK-099402, R01 DK-092606; grants from the American Diabetes Association: 7-12-BS-092, 1-14-Merck-10; a VA Merit Award (I01-BX000901); and by the Novo Nordisk Foundation Center for Basic Metabolic Research. Its contents are solely the responsibility of the authors and do not necessarily represent the official view of the National Center for Research Resources or National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

A.K.M., D.M.E., V.T.S., R.G.K., J.-P.C., M.J.J., H.-Y.L. and G.I.S. designed the experimental protocols. A.K.M., D.M.E., Y.R., J.H., X-M.Z., G.W.C., R.G.K., J.-P.C., M.J.J. and H.-Y.L. performed the studies. A.K.M., D.M.E., X-M.Z., D.B., R.A.A., G.W.C., J.-P.C., M.J.J. and H.-Y.L. analysed the data. B.J.P., J.L.W., S.B. and M.J.M. supplied reagents. A.K.M., D.M.E., Y.R., G.W.C., V.T.S., R.G.K., R.A.A., M.J.M. and G.I.S. contributed to writing the manuscript.

Corresponding author

Correspondence to Gerald I. Shulman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Effect of acute galegine treatment and acute AMPK activator treatment in vivo.

A 20-min infusion of galegine in rats (a) decreased fasting plasma glucose concentrations, (b) decreased fasting plasma insulin concentrations, and (c) increased plasma lactate concentrations, (d) independently of any changes in gluconeogenic gene expression. e, AMPK was activated by acute galegine treatment. f, Twenty-minute infusions of A-769662 in rats had no effect on fasting plasma glucose concentrations or (g) endogenous glucose production, in spite of (h) comparable activation of AMPK. Data are mean ± s.e.m. (saline, n = 6; galegine, n = 8; A-769662, n = 5 biological replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 2 Effect of guanide/biguanide treatment on enzymes involved in pyruvate metabolism, redox regulation and the malate–aspartate shuttle, and on complex-I-mediated respiration.

a, Lactate enters metabolism through LDH by a redox-dependent reaction into the pyruvate pool. Pyruvate lies at the intersection of alanine influx, glycolysis, citric acid cycle and gluconeogenic flux. b, Guanides/biguanides did not affect pyruvate carboxylase (PC) activity, compared with the known inhibitor coenzyme A, and (c) did not affect citrate synthase activity. d, Alanine aminotransferase activity was also unaffected. e, Guanides/biguanides did not affect MDH activity, (f) ASAT activity or (g) total shuttle rates. h, Metformin had no effect on complex-I-mediated respiration in isolated mitochondria at concentrations less than 5 mM, and induced a slight increase in complex II respiration; data shown representative of three experiments. Data are mean ± s.e.m. (n = 5 technical replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 3 Effect of acute metformin (20 mg kg−1 and 50 mg kg−1, intravenously) treatment on EGP, liver redox, energy charge and liver gluconeogenic protein expression in Sprague-Dawley rats.

a, Acute metformin (20 mg kg−1) treatment significantly lowered EGP, (b) increased the liver [lactate]:[:pyruvate] ratio and (c) decreased liver [β-hydroxybutyrate]:[acetoacetate]. d, Acute metformin (50 mg kg−1) treatment increased liver [GSSG]:[GSH], (e) but had no effect on the liver [ATP]:[ADP] or (f) [ATP]:[AMP] ratios. g, The [NADH]:[NAD+] and (h) [NADPH]:[NADP+] ratios also remained unchanged. i, Acute metformin treatment had no effect on liver [cAMP] levels. j, Acute metformin (50 mg kg−1) treatment had no effect on the expression of key gluconeogenic enzymes or AMPK in the liver. k, PEPCK-C protein expression and (l) pyruvate carboxylase protein expression were unchanged, although (m) activated CREB as determined by the ratio of phosphorylated CREB to total CREB levels was slightly increased. n, There was no activation of liver AMPK, as reflected by the ratio of phosphorylated AMPK to total AMPK levels, and (o) no change in the phosphorylation of AMPK downstream target ACC. Data are mean ± s.e.m. (n = 6 biological replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 4 Effect of chronic metformin (50 mg kg−1 per day, intraperitoneally for 30 days) treatment on liver redox, energy charge and expression of gluconeogenic regulators.

a, Chronic metformin treatment increased the liver [GSSG]:[GSH] ratio, but (b) had no effect on the liver [ATP]:[ADP] or (c) [ATP]:[AMP] ratios. d, The [NADH]:[NAD+] and (e) [NADPH]:[NADP+] ratios also remained unchanged. f, Chronic metformin treatment slightly reduced liver [cAMP] levels. g, Chronic metformin treatment had no effect on the protein levels of principal gluconeogenic enzymes in the liver; (h) PEPCK-C protein expression and (i) liver pyruvate carboxylase protein levels both remaining unaltered. j, Activated CREB as determined by the ratio of phosphorylated CREB to total CREB levels was decreased. k, Chronic metformin treatment activated liver AMPK as indicated by the increased ratio of phosphorylated AMPK to total AMPK levels and (l) increased phosphorylation of ACC. Data are mean ± s.e.m. (n = 6 biological replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 5 Effect of metformin on the glycerophosphate shuttle.

NADH made in the cytosol by glycolysis cannot cross the mitochondrial membrane and contribute electrons to the electron transport chain (ETC) for ATP synthesis. Two mechanisms, the reversible malate–aspartate shuttle and (a) the unidirectional glycerophosphate shuttle, oxidize NADH in the cytosol and transport electrons into the mitochondria through metabolic intermediates. The glycerophosphate shuttle is composed of cytosolic and mitochondrial glycerophosphate dehydrogenases, two structurally distinct enzymes. b, Metformin had no effect on cGPD, which consists of two subunits and catalyses the conversion of dihydroyacetone phosphate (DHAP) to glycerol-3-phosphate (G-3-P), oxidizing one NADH. c, Metformin inhibited the activity of rat mGPD, a FAD-linked enzyme that transmits electron pairs to the electron transport chain through the quinone pool, purified from liver by immunoprecipitation. Inhibition of rat mGPD was non-competitive. Data shown are the average of five separate experiments. d, Metformin inhibited pure, recombinant human mGPD non-competitively, and decreased Vmax without affecting the Michaelis constant (Km). Data shown are representative of two experiments. e, Metformin also inhibited the activity of the bacterial mGPD isoform, Pediococcus sp. α-glycerophosphate oxidase, showing non-competitive kinetics. Data are mean ± s.e.m. (n = 4 or 5 technical replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 6 Effect of metformin and knockdown of mGPD by siRNA on glucose production from various substrates in primary hepatocytes, and metformin-mediated increase in glycerol-3-phosphate concentrations.

a, Metformin treatment (100 μM) and siRNA knockdown of mGpd in rat primary hepatocytes inhibited glucose production at higher [lactate]:[pyruvate] ratios, but lower redox state induced by decreased [lactate]:[pyruvate] abrogated the ability of metformin and mGpd knockdown by siRNA to decrease glucose production. Decreasing the redox state itself inhibited glucose production. b, Metformin inhibited glucose production only from lactate and glycerol, not from substrates that did not increase the cytosolic redox state. c, mGpd knockdown by siRNA showed a similar substrate-selective inhibition of glucose production. d, Both metformin and mGPD siRNA treatment increased [glycerol-3-phosphate] levels in hepatocytes, and (e) acute metformin (50 mg kg−1, intravenously) treatment in vivo increased liver [glycerol-3-phosphate] levels without significantly altering [glycerol-3-phosphate] levels in other tissues, suggesting an impasse at the mGPD catalytic step. f, siRNA treatment did not induce cytotoxicity, as determined by trypan blue exclusion, (g) CyQuant proliferation assay and (h) the absence of cytochrome c release into the cytosolic fraction from mitochondria of treated cells. Data are mean ± s.e.m. (n = 5 technical replicates, n = 3 for cytotoxicity tests (fh)). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 7 Effect of mGPD and cGPD ASO treatment on liver redox, high-energy intermediates and expression of gluconeogenic regulators.

a, mGPD ASO effectively reduced expression of liver mGPD protein and (b) cGPD ASO effectively reduced liver cGPD protein levels. c, mGPD ASO treatment increased plasma lactate concentrations significantly, but cGPD ASO knockdown had no effect on plasma lactate concentrations. d, mGPD ASO knockdown increased the liver [GSSG]:[GSH] ratio, (e) had no effect on the liver [ATP]:[ADP], (f) [ATP]:[AMP], (g) [NADH]:[NAD+] or (h) [NADPH]:[NADP+] ratios, (i) although liver [cAMP] levels were slightly decreased. j, ASO-mediated knockdown of mGPD did not affect expression of gluconeogenic enzymes, (k) PEPCK-C and (l) pyruvate carboxylase protein levels remaining unchanged in the liver. m, Activated CREB was decreased, and (n) mGPD ASO knockdown led to activation of liver AMPK as indicated by increased phosphorylated AMPK and (o) increased ACC phosphorylation. Data are mean ± s.e.m. (n = 6 biological replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

Extended Data Figure 8 Computational binding model of guanides/biguanides to mGPD from Streptococcus sp.

Modelling of guanide/biguanide binding to mGPD after modification of key residues to fit the human sequence, show (a) FAD binding and predicted movement in the pocket, (b) metformin binding and (c) phenformin binding to the FAD-containing pocket.

Extended Data Figure 9 Plasma and tissue metformin concentrations in rats treated with metformin.

a, Acute metformin (50 mg kg−1, intravenously) administration led to peak plasma metformin concentrations of approximately 74 μM; 100 and 250 mg kg−1 doses increased plasma metformin concentration to 345 and 1300 μM, respectively. b, Acute metformin (50 mg kg−1, intravenously) led to liver metformin concentrations of approximately 100 μM, and metformin levels in other tissues were comparatively low. Data shown are representative of two experiments. Data are mean ± s.e.m. (n = 3 biological replicates for plasma concentrations, n = 5 biological replicates for tissue levels). *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA.

Extended Data Figure 10 Effect of mGPD and cGPD ASO treatment on liver toxicity and tissue-specific knockdown of mGPD expression by mGPD ASO.

a, mGPD ASO treatment had no effect on body weight after treatment at 37.5 mg kg−1 ASO twice a week for 4 weeks. b, All ASOs screened in this study, mGPD ASO 1, mGPD ASO 2, cGPD ASO 1 and cGPD ASO 2, elicited no significant liver toxicity as determined by plasma AST/ALT levels after 4 weeks. c, Treatment with mGPD ASO 2 for 4 weeks during the mGPD ASO with acute metformin study also had no effect on plasma AST/ALT. d, mGPD ASO treatment led to cleavage of mGPD mRNA transcript exclusively in the liver, only slightly decreasing transcript levels in white adipose tissue and having no effect on mGPD mRNA in other tissues. ek, mGPD ASO treatment specifically reduced protein expression of mGPD in the liver, with no significant effect on mGPD protein levels in the pancreas, kidney, muscle, white adipose tissue or brown adipose tissue. Data are mean ± s.e.m. (n = 6 biological replicates). *P < 0.05, **P < 0.01, ***P < 0.001 by unpaired t-test.

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Madiraju, A., Erion, D., Rahimi, Y. et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 510, 542–546 (2014). https://doi.org/10.1038/nature13270

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13270

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing