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.

Targeting hepatic glutaminase activity to ameliorate hyperglycemia

A Publisher Correction to this article was published on 12 June 2018

This article has been updated

Abstract

Glucagon levels increase under homeostatic, fasting conditions, promoting the release of glucose from the liver by accelerating the breakdown of glycogen (also known as glycogenolysis). Glucagon also enhances gluconeogenic flux, including from an increase in the hepatic consumption of amino acids1. In type 2 diabetes, dysregulated glucagon signaling contributes to the elevated hepatic glucose output and fasting hyperglycemia that occur in this condition. Yet, the mechanism by which glucagon stimulates gluconeogenesis remains incompletely understood. Contrary to the prevailing belief that glucagon acts primarily on cytoplasmic and nuclear targets, we find glucagon-dependent stimulation of mitochondrial anaplerotic flux from glutamine that increases the contribution of this amino acid to the carbons of glucose generated during gluconeogenesis. This enhanced glucose production is dependent on protein kinase A (PKA) and is associated with glucagon-stimulated calcium release from the endoplasmic reticulum, activation of mitochondrial α-ketoglutarate dehydrogenase, and increased glutaminolysis. Mice with reduced levels of hepatic glutaminase 2 (GLS2), the enzyme that catalyzes the first step in glutamine metabolism, show lower glucagon-stimulated glutamine-to-glucose flux in vivo, and GLS2 knockout results in higher fasting plasma glucagon and glutamine levels with lower fasting blood glucose levels in insulin-resistant conditions. As found in genome-wide association studies (GWAS), human genetic variation in the region of GLS2 is associated with higher fasting plasma glucose2,3; here we show in human cryopreserved primary hepatocytes in vitro that these natural gain-of-function missense mutations in GLS2 result in higher glutaminolysis and glucose production. These data emphasize the importance of gluconeogenesis from glutamine, particularly in pathological states of increased glucagon signaling, while suggesting a possible new therapeutic avenue to treat hyperglycemia.

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: Glucagon-mediated metabolic flux studies from glutamine in primary hepatocytes.
Figure 2: Kinetics effects of glucagon on α-ketoglutarate and glutamate in primary hepatocytes.
Figure 3: In vivo glutamine and glucagon infusion study from mice infected with AAV-GFP or AAV-GLS2-sh.
Figure 4: Glutamine metabolism in primary cryopreserved human hepatocytes from donors genotyped to be homozygous (L581L/L581) and heterozygous (L581/P581) at GLS2.

Change history

  • 12 June 2018

    In the version of this article initially published, the "[13C2]α-ketoglutarate" label on Fig. 1g is incorrect. It should be "[13C5]α-ketoglutarate". Additionally, in Fig. 3b, the "AAV-GFP" group is missing a notation for significance, and in Fig. 3c, the "AAV-GLS2-sh" group is missing a notation for significance. There should be a double asterisk notating significance in both panels. Finally, in the Fig. 4g legend, "[13C6]UDP-glucose" should be "[13C3]UDP-glucose", and in the Fig. 4h legend, "[13C6]hexose" should be "[13C3]hexose". The errors have been corrected in the HTML and PDF versions of this article.

References

  1. Miller, R.A. & Birnbaum, M.J. Glucagon: acute actions on hepatic metabolism. Diabetologia 59, 1376–1381 (2016).

    Article  CAS  Google Scholar 

  2. Scott, R.A. et al. Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nat. Genet. 44, 991–1005 (2012).

    Article  CAS  Google Scholar 

  3. Suhre, K. et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature 477, 54–60 (2011).

    Article  CAS  Google Scholar 

  4. Exton, J.H. & Park, C.R. Control of gluconeogenesis in liver. II. Effects of glucagon, catecholamines, and adenosine 3′,5′-monophosphate on gluconeogenesis in the perfused rat liver. J. Biol. Chem. 243, 4189–4196 (1968).

    CAS  PubMed  Google Scholar 

  5. Young, V.R. & Ajami, A.M. Glutamine: the emperor or his clothes? J. Nutr. 131, 2449S–2459S discussion 2486S–2447S (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  7. Titchenell, P.M. et al. Direct hepatocyte insulin signaling is required for lipogenesis but is dispensable for the suppression of glucose production. Cell Metab. 23, 1154–1166 (2016).

    Article  CAS  Google Scholar 

  8. Williamson, J.R. et al. Mechanisms involved in receptor-mediated changes of intracellular Ca2+ in liver. Soc. Gen. Physiol. Ser. 42, 93–116 (1987).

    CAS  PubMed  Google Scholar 

  9. Watford, M. & Smith, E.M. Distribution of hepatic glutaminase activity and mRNA in perivenous and periportal rat hepatocytes. Biochem. J. 267, 265–267 (1990).

    Article  CAS  Google Scholar 

  10. Hu, W. et al. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc. Natl. Acad. Sci. USA 107, 7455–7460 (2010).

    Article  CAS  Google Scholar 

  11. Suzuki, S. et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc. Natl. Acad. Sci. USA 107, 7461–7466 (2010).

    Article  CAS  Google Scholar 

  12. Krishna, M.G. et al. Glucagon response to exercise is critical for accelerated hepatic glutamine metabolism and nitrogen disposal. Am. J. Physiol. Endocrinol. Metab. 279, E638–E645 (2000).

    Article  CAS  Google Scholar 

  13. Stumvoll, M., Meyer, C., Kreider, M., Perriello, G. & Gerich, J. Effects of glucagon on renal and hepatic glutamine gluconeogenesis in normal postabsorptive humans. Metabolism 47, 1227–1232 (1998).

    Article  CAS  Google Scholar 

  14. Battezzati, A., Simonson, D.C., Luzi, L. & Matthews, D.E. Glucagon increases glutamine uptake without affecting glutamine release in humans. Metabolism 47, 713–723 (1998).

    Article  CAS  Google Scholar 

  15. Hankard, R.G., Haymond, M.W. & Darmaun, D. Role of glutamine as a glucose precursor in fasting humans. Diabetes 46, 1535–1541 (1997).

    Article  CAS  Google Scholar 

  16. Stumvoll, M. et al. Glutamine and alanine metabolism in NIDDM. Diabetes 45, 863–868 (1996).

    Article  Google Scholar 

  17. Perriello, G. et al. Estimation of glucose–alanine–lactate–glutamine cycles in postabsorptive humans: role of skeletal muscle. Am. J. Physiol. 269, E443–E450 (1995).

    Article  CAS  Google Scholar 

  18. Nurjhan, N. et al. Glutamine: a major gluconeogenic precursor and vehicle for interorgan carbon transport in man. J. Clin. Invest. 95, 272–277 (1995).

    Article  CAS  Google Scholar 

  19. Halestrap, A.P. Stimulation of the respiratory chain of rat liver mitochondria between cytochrome c1 and cytochrome c by glucagon treatment of rats. Biochem. J. 172, 399–405 (1978).

    Article  CAS  Google Scholar 

  20. Yamazaki, R.K. Glucagon stimulation of mitochondrial respiration. J. Biol. Chem. 250, 7924–7930 (1975).

    CAS  PubMed  Google Scholar 

  21. Stumvoll, M., Perriello, G., Meyer, C. & Gerich, J. Role of glutamine in human carbohydrate metabolism in kidney and other tissues. Kidney Int. 55, 778–792 (1999).

    Article  CAS  Google Scholar 

  22. Mauger, J.P., Claret, M., Pietri, F. & Hilly, M. Hormonal regulation of inositol 1,4,5-trisphosphate receptor in rat liver. J. Biol. Chem. 264, 8821–8826 (1989).

    CAS  PubMed  Google Scholar 

  23. Burgess, G.M., Bird, G.S., Obie, J.F. & Putney, J.W. Jr. The mechanism for synergism between phospholipase C– and adenylylcyclase-linked hormones in liver. Cyclic AMP–dependent kinase augments inositol trisphosphate–mediated Ca2+ mobilization without increasing the cellular levels of inositol polyphosphates. J. Biol. Chem. 266, 4772–4781 (1991).

    CAS  PubMed  Google Scholar 

  24. Bygrave, F.L., Gamberucci, A., Fulceri, R. & Benedetti, A. Evidence that stimulation of plasma-membrane Ca2+ inflow is an early action of glucagon and dibutyryl cyclic AMP in rat hepatocytes. Biochem. J. 292, 19–22 (1993).

    Article  CAS  Google Scholar 

  25. Fernando, K.C., Gregory, R.B. & Barritt, G.J. Protein kinase A regulates the disposition of Ca2+ which enters the cytoplasmic space through store-activated Ca2+ channels in rat hepatocytes by diverting inflowing Ca2+ to mitochondria. Biochem. J. 330, 1179–1187 (1998).

    Article  CAS  Google Scholar 

  26. Hughes, B.P. & Barritt, G.J. Effects of glucagon and N6O2′-dibutyryladenosine 3′:5′-cyclic monophosphate on calcium transport in isolated rat liver mitochondria. Biochem. J. 176, 295–304 (1978).

    Article  CAS  Google Scholar 

  27. Keppens, S., Vandenheede, J.R. & De Wulf, H. On the role of calcium as second messenger in liver for the hormonally induced activation of glycogen phosphorylase. Biochim. Biophys. Acta 496, 448–457 (1977).

    Article  CAS  Google Scholar 

  28. Denton, R.M. & McCormack, J.G. Ca2+ transport by mammalian mitochondria and its role in hormone action. Am. J. Physiol. 249, E543–E554 (1985).

    CAS  PubMed  Google Scholar 

  29. Siess, E.A. & Wieland, O.H. Glucagon-induced stimulation of 2-oxoglutarate metabolism in mitochondria from rat liver. FEBS Lett. 93, 301–306 (1978).

    Article  CAS  Google Scholar 

  30. Williamson, J.R., Browning, E.T., Thurman, R.G. & Scholz, R. Inhibition of glucagon effects in perfused rat liver by (+)decanoylcarnitine. J. Biol. Chem. 244, 5055–5064 (1969).

    CAS  PubMed  Google Scholar 

  31. Lacey, J.H., Bradford, N.M., Joseph, S.K. & McGivan, J.D. Increased activity of phosphate-dependent glutaminase in liver mitochondria as a result of glucagon treatment of rats. Biochem. J. 194, 29–33 (1981).

    Article  CAS  Google Scholar 

  32. Joseph, S.K. & McGivan, J.D. The effect of ammonium chloride and glucagon on the metabolism of glutamine in isolated liver cells from starved rats. Biochim. Biophys. Acta 543, 16–28 (1978).

    Article  CAS  Google Scholar 

  33. Low, S.Y., Salter, M., Knowles, R.G., Pogson, C.I. & Rennie, M.J. A quantitative analysis of the control of glutamine catabolism in rat liver cells. Use of selective inhibitors. Biochem. J. 295 (Pt. 2), 617–624 (1993).

    Article  CAS  Google Scholar 

  34. Lu, W. et al. Metabolomic analysis via reversed-phase ion-pairing liquid chromatography coupled to a stand alone orbitrap mass spectrometer. Anal. Chem. 82, 3212–3221 (2010).

    Article  CAS  Google Scholar 

  35. Melamud, E., Vastag, L. & Rabinowitz, J.D. Metabolomic analysis and visualization engine for LC–MS data. Anal. Chem. 82, 9818–9826 (2010).

    Article  CAS  Google Scholar 

  36. Weitzel, M. et al. 13CFLUX2—high-performance software suite for 13C-metabolic flux analysis. Bioinformatics 29, 143–145 (2013).

    Article  CAS  Google Scholar 

  37. Wiechert, W., Mollney, M., Isermann, N., Wurzel, M. & de Graaf, A.A. Bidirectional reaction steps in metabolic networks: III. Explicit solution and analysis of isotopomer labeling systems. Biotechnol. Bioeng. 66, 69–85 (1999).

    Article  CAS  Google Scholar 

  38. Antoniewicz, M.R., Kelleher, J.K. & Stephanopoulos, G. Determination of confidence intervals of metabolic fluxes estimated from stable isotope measurements. Metab. Eng. 8, 324–337 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

All data generated and analyzed within this study are presented in the article and supplementary procedures. The stable-isotope-based metabolomics work was supported by US NIH grant CA211437 to W.L. This work was also supported by FONDECYT grant 1160332 to C.C. and CONICYT/FONDAP 15150012 to C.C.

Author information

Authors and Affiliations

Authors

Contributions

R.A.M. designed and performed all experiments and drafted and edited the manuscript. Y.S. designed and performed experiments related to GLS2 knockout and in vivo infusion and drafted and edited the manuscript. W.L. performed mass spectrometry experiments in rodent cells and tissues and edited the manuscript. D.A.P. performed mass spectrometry experiments in human hepatocytes and edited the manuscript. A.J. performed experiments on human hepatocytes and edited the manuscript. M.B. performed mass spectrometry experiments in human hepatocytes and edited the manuscript. H.W. performed experiments on human hepatocytes and edited the manuscript. C.C. performed mouse hepatocyte calcium experiments and edited the manuscript. M.W. performed experiments related to GLS2 knockout and in vivo infusions and edited the manuscript. J.K.F. edited the manuscript. J.O.P. performed flux modeling experiments and edited the manuscript. Y.Z. performed pancreas histology experiments and edited the manuscript. W.L.H. designed pancreas histology experiments and edited the manuscript. J.D.R. designed experiments and drafted and edited the manuscript. M.J.B. designed experiments and drafted and edited the manuscript.

Corresponding authors

Correspondence to Russell A Miller or Morris J Birnbaum.

Ethics declarations

Competing interests

R.A.M., Y.S., D.A.P., A.J., M.B., H.W., M.W., and M.J.B. were employed by Pfizer during the reported studies.

Supplementary information

Supplementary Figures & Tables

Supplementary Figures 1–7 & Supplementary Table 1 (PDF 1428 kb)

Life Sciences Reporting Summary (PDF 130 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miller, R., Shi, Y., Lu, W. et al. Targeting hepatic glutaminase activity to ameliorate hyperglycemia. Nat Med 24, 518–524 (2018). https://doi.org/10.1038/nm.4514

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4514

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research