Skip to main content
Top
Published in: Diabetologia 10/2017

01-10-2017 | Article

Mitochondrial H+-ATP synthase in human skeletal muscle: contribution to dyslipidaemia and insulin resistance

Authors: Laura Formentini, Alexander J. Ryan, Manuel Gálvez-Santisteban, Leslie Carter, Pam Taub, John D. Lapek Jr., David J. Gonzalez, Francisco Villarreal, Theodore P. Ciaraldi, José M. Cuezva, Robert R. Henry

Published in: Diabetologia | Issue 10/2017

Login to get access

Abstract

Aims/hypothesis

Mitochondria are important regulators of the metabolic phenotype in type 2 diabetes. A key factor in mitochondrial physiology is the H+-ATP synthase. The expression and activity of its physiological inhibitor, ATPase inhibitory factor 1 (IF1), controls tissue homeostasis, metabolic reprogramming and signalling. We aimed to characterise the putative role of IF1 in mediating skeletal muscle metabolism in obesity and diabetes.

Methods

We examined the ‘mitochondrial signature’ of obesity and type 2 diabetes in a cohort of 100 metabolically characterised human skeletal muscle biopsy samples. The expression and activity of H+-ATP synthase, IF1 and key mitochondrial proteins were characterised, including their association with BMI, fasting plasma insulin, fasting plasma glucose and HOMA-IR. IF1 was also overexpressed in primary cultures of human myotubes derived from the same biopsies to unveil the possible role played by the pathological inhibition of the H+-ATP synthase in skeletal muscle.

Results

The results indicate that type 2 diabetes and obesity act via different mechanisms to impair H+-ATP synthase activity in human skeletal muscle (76% reduction in its catalytic subunit vs 280% increase in IF1 expression, respectively) and unveil a new pathway by which IF1 influences lipid metabolism. Mechanistically, IF1 altered cellular levels of α-ketoglutarate and l-carnitine metabolism in the myotubes of obese (84% of control) and diabetic (76% of control) individuals, leading to limited β-oxidation of fatty acids (60% of control) and their cytosolic accumulation (164% of control). These events led to enhanced release of TNF-α (10 ± 2 pg/ml, 27 ± 5 pg/ml and 35 ± 4 pg/ml in control, obese and type 2 diabetic participants, respectively), which probably contributes to an insulin resistant phenotype.

Conclusions/interpretation

Overall, our data highlight IF1 as a novel regulator of lipid metabolism and metabolic disorders, and a possible target for therapeutic intervention.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lowell BB, Shulman GI (2005) Mitochondrial dysfunction and type 2 diabetes. Science 307:384–387CrossRef Lowell BB, Shulman GI (2005) Mitochondrial dysfunction and type 2 diabetes. Science 307:384–387CrossRef
2.
go back to reference Sivitz WI, Yorek MA (2010) Mitochondrial dysfunction in diabetes: from molecular mechanisms to functional significance and therapeutic opportunities. Antioxid Redox Signal 12:537–577CrossRef Sivitz WI, Yorek MA (2010) Mitochondrial dysfunction in diabetes: from molecular mechanisms to functional significance and therapeutic opportunities. Antioxid Redox Signal 12:537–577CrossRef
3.
go back to reference Asmann YW, Stump CS, Short KR et al (2006) Skeletal muscle mitochondrial functions, mitochondrial DNA copy numbers, and gene transcript profiles in type 2 diabetic and nondiabetic subjects at equal levels of low or high insulin and euglycemia. Diabetes 55:3309–3319CrossRef Asmann YW, Stump CS, Short KR et al (2006) Skeletal muscle mitochondrial functions, mitochondrial DNA copy numbers, and gene transcript profiles in type 2 diabetic and nondiabetic subjects at equal levels of low or high insulin and euglycemia. Diabetes 55:3309–3319CrossRef
4.
go back to reference Sanchez-Arago M, Formentini L, Cuezva JM (2013) Mitochondria-mediated energy adaption in cancer: the H(+)-ATP synthase-geared switch of metabolism in human tumors. Antioxid Redox Signal 19:285–298CrossRef Sanchez-Arago M, Formentini L, Cuezva JM (2013) Mitochondria-mediated energy adaption in cancer: the H(+)-ATP synthase-geared switch of metabolism in human tumors. Antioxid Redox Signal 19:285–298CrossRef
5.
go back to reference Friedman JR, Nunnari J (2014) Mitochondrial form and function. Nature 505:335–343CrossRef Friedman JR, Nunnari J (2014) Mitochondrial form and function. Nature 505:335–343CrossRef
6.
go back to reference Yoshida M, Muneyuki E, Hisabori T (2001) ATP synthase—a marvellous rotary engine of the cell. Nat Rev Mol Cell Biol 2:669–677CrossRef Yoshida M, Muneyuki E, Hisabori T (2001) ATP synthase—a marvellous rotary engine of the cell. Nat Rev Mol Cell Biol 2:669–677CrossRef
7.
go back to reference Formentini L, Pereira MP, Sanchez-Cenizo L et al (2014) In vivo inhibition of the mitochondrial H+-ATP synthase in neurons promotes metabolic preconditioning. EMBO J 33:762–778CrossRef Formentini L, Pereira MP, Sanchez-Cenizo L et al (2014) In vivo inhibition of the mitochondrial H+-ATP synthase in neurons promotes metabolic preconditioning. EMBO J 33:762–778CrossRef
8.
go back to reference Strauss M, Hofhaus G, Schroder RR, Kuhlbrandt W (2008) Dimer ribbons of ATP synthase shape the inner mitochondrial membrane. EMBO J 27:1154–1160CrossRef Strauss M, Hofhaus G, Schroder RR, Kuhlbrandt W (2008) Dimer ribbons of ATP synthase shape the inner mitochondrial membrane. EMBO J 27:1154–1160CrossRef
9.
go back to reference Formentini L, Sanchez-Arago M, Sanchez-Cenizo L, Cuezva JM (2012) The mitochondrial ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative response. Mol Cell 45:731–742CrossRef Formentini L, Sanchez-Arago M, Sanchez-Cenizo L, Cuezva JM (2012) The mitochondrial ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative response. Mol Cell 45:731–742CrossRef
10.
go back to reference Martinez-Reyes I, Cuezva JM (2014) The H(+)-ATP synthase: a gate to ROS-mediated cell death or cell survival. Biochim Biophys Acta 1837:1099–1112CrossRef Martinez-Reyes I, Cuezva JM (2014) The H(+)-ATP synthase: a gate to ROS-mediated cell death or cell survival. Biochim Biophys Acta 1837:1099–1112CrossRef
11.
go back to reference Hojlund K, Wrzesinski K, Larsen PM et al (2003) Proteome analysis reveals phosphorylation of ATP synthase beta -subunit in human skeletal muscle and proteins with potential roles in type 2 diabetes. J Biol Chem 278:10436–10442CrossRef Hojlund K, Wrzesinski K, Larsen PM et al (2003) Proteome analysis reveals phosphorylation of ATP synthase beta -subunit in human skeletal muscle and proteins with potential roles in type 2 diabetes. J Biol Chem 278:10436–10442CrossRef
12.
go back to reference Guan SS, Sheu ML, Wu CT, Chiang CK, Liu SH (2015) ATP synthase subunit-beta down-regulation aggravates diabetic nephropathy. Sci Rep 5:14561CrossRef Guan SS, Sheu ML, Wu CT, Chiang CK, Liu SH (2015) ATP synthase subunit-beta down-regulation aggravates diabetic nephropathy. Sci Rep 5:14561CrossRef
13.
go back to reference Hojlund K, Yi Z, Lefort N et al (2010) Human ATP synthase beta is phosphorylated at multiple sites and shows abnormal phosphorylation at specific sites in insulin-resistant muscle. Diabetologia 53:541–551CrossRef Hojlund K, Yi Z, Lefort N et al (2010) Human ATP synthase beta is phosphorylated at multiple sites and shows abnormal phosphorylation at specific sites in insulin-resistant muscle. Diabetologia 53:541–551CrossRef
14.
go back to reference Garcia-Bermudez J, Cuezva JM (2016) The ATPase inhibitory factor 1 (IF1): a master regulator of energy metabolism and of cell survival. Biochim Biophys Acta 1857:1167–1182CrossRef Garcia-Bermudez J, Cuezva JM (2016) The ATPase inhibitory factor 1 (IF1): a master regulator of energy metabolism and of cell survival. Biochim Biophys Acta 1857:1167–1182CrossRef
15.
go back to reference Gledhill JR, Montgomery MG, Leslie AG, Walker JE (2007) How the regulatory protein, IF(1), inhibits F(1)-ATPase from bovine mitochondria. Proc Natl Acad Sci U S A 104:15671–15676CrossRef Gledhill JR, Montgomery MG, Leslie AG, Walker JE (2007) How the regulatory protein, IF(1), inhibits F(1)-ATPase from bovine mitochondria. Proc Natl Acad Sci U S A 104:15671–15676CrossRef
16.
go back to reference Sanchez-Arago M, Formentini L, Martinez-Reyes I et al (2013) Expression, regulation and clinical relevance of the ATPase inhibitory factor 1 in human cancers. Oncogenesis 2:e46CrossRef Sanchez-Arago M, Formentini L, Martinez-Reyes I et al (2013) Expression, regulation and clinical relevance of the ATPase inhibitory factor 1 in human cancers. Oncogenesis 2:e46CrossRef
17.
go back to reference Sanchez-Arago M, Garcia-Bermudez J, Martinez-Reyes I, Santacatterina F, Cuezva JM (2013) Degradation of IF1 controls energy metabolism during osteogenic differentiation of stem cells. EMBO Rep 14:638–644CrossRef Sanchez-Arago M, Garcia-Bermudez J, Martinez-Reyes I, Santacatterina F, Cuezva JM (2013) Degradation of IF1 controls energy metabolism during osteogenic differentiation of stem cells. EMBO Rep 14:638–644CrossRef
18.
go back to reference Vazquez-Martin A, Corominas-Faja B, Cufi S et al (2013) The mitochondrial H(+)-ATP synthase and the lipogenic switch: new core components of metabolic reprogramming in induced pluripotent stem (iPS) cells. Cell Cycle 12:207–218CrossRef Vazquez-Martin A, Corominas-Faja B, Cufi S et al (2013) The mitochondrial H(+)-ATP synthase and the lipogenic switch: new core components of metabolic reprogramming in induced pluripotent stem (iPS) cells. Cell Cycle 12:207–218CrossRef
19.
go back to reference Henry RR, Abrams L, Nikoulina S, Ciaraldi TP (1995) Insulin action and glucose metabolism in nondiabetic control and NIDDM subjects. Comparison using human skeletal muscle cell cultures. Diabetes 44:936–946CrossRef Henry RR, Abrams L, Nikoulina S, Ciaraldi TP (1995) Insulin action and glucose metabolism in nondiabetic control and NIDDM subjects. Comparison using human skeletal muscle cell cultures. Diabetes 44:936–946CrossRef
20.
go back to reference Ciaraldi TP, Abrams L, Nikoulina S, Mudaliar S, Henry RR (1995) Glucose transport in cultured human skeletal muscle cells. Regulation by insulin and glucose in nondiabetic and non-insulin-dependent diabetes mellitus subjects. J Clin Invest 96:2820–2827CrossRef Ciaraldi TP, Abrams L, Nikoulina S, Mudaliar S, Henry RR (1995) Glucose transport in cultured human skeletal muscle cells. Regulation by insulin and glucose in nondiabetic and non-insulin-dependent diabetes mellitus subjects. J Clin Invest 96:2820–2827CrossRef
21.
go back to reference Santacatterina F, Sanchez-Cenizo L, Formentini L et al (2016) Down-regulation of oxidative phosphorylation in the liver by expression of the ATPase inhibitory factor 1 induces a tumor-promoter metabolic state. Oncotarget 7:490–508CrossRef Santacatterina F, Sanchez-Cenizo L, Formentini L et al (2016) Down-regulation of oxidative phosphorylation in the liver by expression of the ATPase inhibitory factor 1 induces a tumor-promoter metabolic state. Oncotarget 7:490–508CrossRef
22.
go back to reference Cha BS, Ciaraldi TP, Carter L et al (2001) Peroxisome proliferator-activated receptor (PPAR) gamma and retinoid X receptor (RXR) agonists have complementary effects on glucose and lipid metabolism in human skeletal muscle. Diabetologia 44:444–452CrossRef Cha BS, Ciaraldi TP, Carter L et al (2001) Peroxisome proliferator-activated receptor (PPAR) gamma and retinoid X receptor (RXR) agonists have complementary effects on glucose and lipid metabolism in human skeletal muscle. Diabetologia 44:444–452CrossRef
23.
go back to reference Wang Y, Yang F, Gritsenko MA et al (2011) Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics 11:2019–2026CrossRef Wang Y, Yang F, Gritsenko MA et al (2011) Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics 11:2019–2026CrossRef
24.
go back to reference Garcia-Bermudez J, Sanchez-Arago M, Soldevilla B, del Arco A, Nuevo-Tapioles C, Cuezva JM (2015) PKA phosphorylates the ATPase inhibitory factor 1 and inactivates its capacity to bind and inhibit the mitochondrial H(+)-ATP synthase. Cell Rep 12:2143–2155CrossRef Garcia-Bermudez J, Sanchez-Arago M, Soldevilla B, del Arco A, Nuevo-Tapioles C, Cuezva JM (2015) PKA phosphorylates the ATPase inhibitory factor 1 and inactivates its capacity to bind and inhibit the mitochondrial H(+)-ATP synthase. Cell Rep 12:2143–2155CrossRef
25.
go back to reference Pernas L, Scorrano L (2016) Mito-morphosis: mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 78:505–531CrossRef Pernas L, Scorrano L (2016) Mito-morphosis: mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu Rev Physiol 78:505–531CrossRef
26.
go back to reference Yang X, Pratley RE, Tokraks S, Bogardus C, Permana PA (2002) Microarray profiling of skeletal muscle tissues from equally obese, non-diabetic insulin-sensitive and insulin-resistant Pima Indians. Diabetologia 45:1584–1593CrossRef Yang X, Pratley RE, Tokraks S, Bogardus C, Permana PA (2002) Microarray profiling of skeletal muscle tissues from equally obese, non-diabetic insulin-sensitive and insulin-resistant Pima Indians. Diabetologia 45:1584–1593CrossRef
27.
go back to reference Reuter SE, Evans AM (2012) Carnitine and acylcarnitines: pharmacokinetic, pharmacological and clinical aspects. Clin Pharmacokinet 51:553–572CrossRef Reuter SE, Evans AM (2012) Carnitine and acylcarnitines: pharmacokinetic, pharmacological and clinical aspects. Clin Pharmacokinet 51:553–572CrossRef
28.
go back to reference Lorenzo M, Fernandez-Veledo S, Vila-Bedmar R, Garcia-Guerra L, De Alvaro C, Nieto-Vazquez I (2008) Insulin resistance induced by tumor necrosis factor-alpha in myocytes and brown adipocytes. J Anim Sci 86:E94–E104CrossRef Lorenzo M, Fernandez-Veledo S, Vila-Bedmar R, Garcia-Guerra L, De Alvaro C, Nieto-Vazquez I (2008) Insulin resistance induced by tumor necrosis factor-alpha in myocytes and brown adipocytes. J Anim Sci 86:E94–E104CrossRef
29.
go back to reference Chouchani ET, Kazak L, Jedrychowski MP et al (2016) Mitochondrial ROS regulate thermogenic energy expenditure and sulfenylation of UCP1. Nature 532:112–116CrossRef Chouchani ET, Kazak L, Jedrychowski MP et al (2016) Mitochondrial ROS regulate thermogenic energy expenditure and sulfenylation of UCP1. Nature 532:112–116CrossRef
30.
go back to reference Sena LA, Chandel NS (2012) Physiological roles of mitochondrial reactive oxygen species. Mol Cell 48:158–167CrossRef Sena LA, Chandel NS (2012) Physiological roles of mitochondrial reactive oxygen species. Mol Cell 48:158–167CrossRef
31.
go back to reference Muoio DM, Newgard CB (2008) Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev Mol Cell Biol 9:193–205CrossRef Muoio DM, Newgard CB (2008) Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev Mol Cell Biol 9:193–205CrossRef
32.
go back to reference Cruz-Topete D, List EO, Okada S, Kelder B, Kopchick JJ (2011) Proteomic changes in the heart of diet-induced pre-diabetic mice. J Proteome 74:716–727CrossRef Cruz-Topete D, List EO, Okada S, Kelder B, Kopchick JJ (2011) Proteomic changes in the heart of diet-induced pre-diabetic mice. J Proteome 74:716–727CrossRef
33.
go back to reference Stump CS, Short KR, Bigelow ML, Schimke JM, Nair KS (2003) Effect of insulin on human skeletal muscle mitochondrial ATP production, protein synthesis, and mRNA transcripts. Proc Natl Acad Sci U S A 100:7996–8001CrossRef Stump CS, Short KR, Bigelow ML, Schimke JM, Nair KS (2003) Effect of insulin on human skeletal muscle mitochondrial ATP production, protein synthesis, and mRNA transcripts. Proc Natl Acad Sci U S A 100:7996–8001CrossRef
34.
go back to reference Pagel-Langenickel I, Bao J, Joseph JJ et al (2008) PGC-1alpha integrates insulin signaling, mitochondrial regulation, and bioenergetic function in skeletal muscle. J Biol Chem 283:22464–22472CrossRef Pagel-Langenickel I, Bao J, Joseph JJ et al (2008) PGC-1alpha integrates insulin signaling, mitochondrial regulation, and bioenergetic function in skeletal muscle. J Biol Chem 283:22464–22472CrossRef
35.
go back to reference Handschin C, Spiegelman BM (2008) The role of exercise and PGC1alpha in inflammation and chronic disease. Nature 454:463–469CrossRef Handschin C, Spiegelman BM (2008) The role of exercise and PGC1alpha in inflammation and chronic disease. Nature 454:463–469CrossRef
36.
go back to reference Shinoda K, Ohyama K, Hasegawa Y et al (2015) Phosphoproteomics identifies CK2 as a negative regulator of beige adipocyte thermogenesis and energy expenditure. Cell Metab 22:997–1008CrossRef Shinoda K, Ohyama K, Hasegawa Y et al (2015) Phosphoproteomics identifies CK2 as a negative regulator of beige adipocyte thermogenesis and energy expenditure. Cell Metab 22:997–1008CrossRef
37.
go back to reference Sullivan LB, Gui DY, Hosios AM, Bush LN, Freinkman E, Vander Heiden MG (2015) Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162:552–563CrossRef Sullivan LB, Gui DY, Hosios AM, Bush LN, Freinkman E, Vander Heiden MG (2015) Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162:552–563CrossRef
38.
go back to reference Titov DV, Cracan V, Goodman RP, Peng J, Grabarek Z, Mootha VK (2016) Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352:231–235CrossRef Titov DV, Cracan V, Goodman RP, Peng J, Grabarek Z, Mootha VK (2016) Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352:231–235CrossRef
39.
go back to reference Chin RM, Fu X, Pai MY et al (2014) The metabolite alpha-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature 510:397–401CrossRef Chin RM, Fu X, Pai MY et al (2014) The metabolite alpha-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature 510:397–401CrossRef
40.
go back to reference Gaster M, Rustan AC, Aas V, Beck-Nielsen H (2004) Reduced lipid oxidation in skeletal muscle from type 2 diabetic subjects may be of genetic origin: evidence from cultured myotubes. Diabetes 53:542–548CrossRef Gaster M, Rustan AC, Aas V, Beck-Nielsen H (2004) Reduced lipid oxidation in skeletal muscle from type 2 diabetic subjects may be of genetic origin: evidence from cultured myotubes. Diabetes 53:542–548CrossRef
41.
go back to reference Kim HE, Grant AR, Simic MS et al (2016) Lipid biosynthesis coordinates a mitochondrial-to-cytosolic stress response. Cell 166:1539–1552CrossRef Kim HE, Grant AR, Simic MS et al (2016) Lipid biosynthesis coordinates a mitochondrial-to-cytosolic stress response. Cell 166:1539–1552CrossRef
42.
go back to reference Febbraio MA, Steensberg A, Starkie RL, McConell GK, Kingwell BA (2003) Skeletal muscle interleukin-6 and tumor necrosis factor-α release in healthy subjects and patients with type 2 diabetes at rest and during exercise. Metabolism 52:939–944CrossRef Febbraio MA, Steensberg A, Starkie RL, McConell GK, Kingwell BA (2003) Skeletal muscle interleukin-6 and tumor necrosis factor-α release in healthy subjects and patients with type 2 diabetes at rest and during exercise. Metabolism 52:939–944CrossRef
43.
go back to reference Feldstein AE, Werneburg NW, Canbay A et al (2004) Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology 40:185–194CrossRef Feldstein AE, Werneburg NW, Canbay A et al (2004) Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology 40:185–194CrossRef
44.
go back to reference Pedersen BK, Febbraio MA (2012) Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol 8:457–465CrossRef Pedersen BK, Febbraio MA (2012) Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol 8:457–465CrossRef
45.
go back to reference Amir Levy Y, Ciaraldi TP, Mudaliar SR, Phillips SA, Henry RR (2015) Excessive secretion of IL-8 by skeletal muscle in type 2 diabetes impairs tube growth: potential role of PI3K and the Tie2 receptor. Am J Physiol Endocrinol Metab 309:E22–E34CrossRef Amir Levy Y, Ciaraldi TP, Mudaliar SR, Phillips SA, Henry RR (2015) Excessive secretion of IL-8 by skeletal muscle in type 2 diabetes impairs tube growth: potential role of PI3K and the Tie2 receptor. Am J Physiol Endocrinol Metab 309:E22–E34CrossRef
46.
go back to reference Kelley DE, He J, Menshikova EV, Ritov VB (2002) Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51:2944–2950CrossRef Kelley DE, He J, Menshikova EV, Ritov VB (2002) Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51:2944–2950CrossRef
47.
go back to reference Morino K, Petersen KF, Dufour S et al (2005) Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest 115:3587–3593CrossRef Morino K, Petersen KF, Dufour S et al (2005) Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents. J Clin Invest 115:3587–3593CrossRef
48.
go back to reference Eckel RH, Kahn SE, Ferrannini E et al (2011) Obesity and type 2 diabetes: what can be unified and what needs to be individualized? Diabetes Care 34:1424–1430CrossRef Eckel RH, Kahn SE, Ferrannini E et al (2011) Obesity and type 2 diabetes: what can be unified and what needs to be individualized? Diabetes Care 34:1424–1430CrossRef
Metadata
Title
Mitochondrial H+-ATP synthase in human skeletal muscle: contribution to dyslipidaemia and insulin resistance
Authors
Laura Formentini
Alexander J. Ryan
Manuel Gálvez-Santisteban
Leslie Carter
Pam Taub
John D. Lapek Jr.
David J. Gonzalez
Francisco Villarreal
Theodore P. Ciaraldi
José M. Cuezva
Robert R. Henry
Publication date
01-10-2017
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 10/2017
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-017-4379-z

Other articles of this Issue 10/2017

Diabetologia 10/2017 Go to the issue