Dysregulation of Glucagon Secretion by Hyperglycemia-Induced Sodium-Dependent Reduction of ATP Production

Jakob G Knudsen, Alexander Hamilton, Reshma Ramracheya, Andrei I Tarasov, Melissa Brereton, Elizabeth Haythorne, Margarita V Chibalina, Peter Spégel, Hindrik Mulder, Quan Zhang, Frances M Ashcroft, Julie Adam, Patrik Rorsman, Jakob G Knudsen, Alexander Hamilton, Reshma Ramracheya, Andrei I Tarasov, Melissa Brereton, Elizabeth Haythorne, Margarita V Chibalina, Peter Spégel, Hindrik Mulder, Quan Zhang, Frances M Ashcroft, Julie Adam, Patrik Rorsman

Abstract

Diabetes is a bihormonal disorder resulting from combined insulin and glucagon secretion defects. Mice lacking fumarase (Fh1) in their β cells (Fh1βKO mice) develop progressive hyperglycemia and dysregulated glucagon secretion similar to that seen in diabetic patients (too much at high glucose and too little at low glucose). The glucagon secretion defects are corrected by low concentrations of tolbutamide and prevented by the sodium-glucose transport (SGLT) inhibitor phlorizin. These data link hyperglycemia, intracellular Na+ accumulation, and acidification to impaired mitochondrial metabolism, reduced ATP production, and dysregulated glucagon secretion. Protein succination, reflecting reduced activity of fumarase, is observed in α cells from hyperglycemic Fh1βKO and β-V59M gain-of-function KATP channel mice, diabetic Goto-Kakizaki rats, and patients with type 2 diabetes. Succination is also observed in renal tubular cells and cardiomyocytes from hyperglycemic Fh1βKO mice, suggesting that the model can be extended to other SGLT-expressing cells and may explain part of the spectrum of diabetic complications.

Keywords: Fh1; diabetes; glucagon; sodium-glucose co-transport; succination.

Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Dysregulation of Glucagon Secretion in Fh1βKO Mice (A) Glucagon secretion in isolated islets from control (CTL; black) and normoglycemic (plasma glucose: 20 mM; red) Fh1βKO mice at 1 and 6 mM glucose. ∗p < 0.05 versus 1 mM glucose; #p < 0.05 versus 1 mM glucose in normoglycemic Fh1βKO islets (n = 8–9 experiments using islets from 12 mice). (B) Islet glucagon content in normoglycemic and hyperglycemic Fh1βKO mice. ∗p < 0.05 (n = 12 mice of each group, each measurement based on 12 islets). (C) Immunohistochemistry (IHC) for succination (2SC) in CTL and Fh1βKO islets. Scale bar, 50 μm. (D) Plasma fumarate levels in CTL and severely hyperglycemic (>20 mM) Fh1βKO mice (n = 22 CTL and n = 13 Fh1βKO mice). (E and F) Glucagon secretion in isolated islets from wild-type (NMRI) islets at 1 and 20 mM glucose and supplementing the extracellular medium with 5 mM Na2-fumarate (E; n = 4 experiments using islets from three mice), or 5 mM dimethyl (dm)-fumarate (F; n = 12 experiments using islets from four mice). ∗p < 0.05 versus 1 mM glucose; #p < 0.05 versus 20 mM glucose. All data presented as mean values ± SEM of indicated number of experiments. See also Figure S1.
Figure 2
Figure 2
Ablation of Fh1 in α Cells Recapitulates the Effects of Diabetes on Glucagon Secretion (A) IHC for 2SC (green), glucagon (red) and overlay (yellow) islets from CTL (above) and Fh1αKO mice. Note strong 2SC labeling of most glucagon-positive cells. Scale bar, 50 μm. Data are representative of four mice of each genotype. (B) Glucagon secretion in CTL and Fh1αKO islets measured during 1 hr static incubations at 1 and 6 mM glucose. ∗p < 0.05 versus 1 mM glucose; #p < 0.05 versus corresponding group in CTL mice (n = 7–8 experiments using islets from three CTL and three Fh1αKO mice). Inset: glucagon secretion at 1 and 6 mM glucose after compensating for α cells that retain Fh1. (C) Glucagon content in islets from CTL and Fh1αKO mice. ∗p < 0.05 (n = 3 mice for each genotype). Inset: glucagon content after compensating for α cells that retain Fh1. (D) Electron micrographs of α cells in a CTL islet (left), a normal α cell in an Fh1αKO islet (center) and an abnormal α cell in an Fh1αKO islet (right; n = 5 cells). Scale bar, 2 μm. All data presented as mean values ± SEM of indicated number of experiments. See also Figure S2.
Figure 3
Figure 3
Hyperglycemia-Induced Changes in Glucagon Secretion Are Corrected by Modulation of KATP Channels (A) IHC for succination (2SC) in CTL and βV59M islets. Scale bar, 50 μm. (B and C) Glucagon secretion in hyperglycemic Fh1βKO (B) and βV59M (C) islets measured during 1 hr static incubations at 1 and 6 mM glucose in the absence or presence of a low concentration (10 μM) of the KATP channel blocker tolbutamide. ∗p < 0.05 versus 1 mM glucose (n = 7–8 experiments using islets from at least three Fh1βKO and three βV59M mice). (D) As in (B) and (C), but experiments obtained from CTL littermates of (n = 6 experiments/6 mice) Fh1βKO and βV59M mice (n = 6–12 experiments/6 mice). ∗p < 0.05 versus 1 mM glucose, #p < 0.05 versus 1 mM glucose without tolbutamide. (E) [Ca2+]i measurements in α cells, using the calcium dye fluo4, within CTL (black) or hyperglycemic Fh1βKO (red) islets at 1 and 20 mM glucose as indicated. In both genotypes α cells were identified by the [Ca2+]i response to adrenaline. (F) Histogram summarizing effects of increasing glucose on [Ca2+]i, measured using fluo4, in α cells from normoglycemic CTL (black) and hyperglycemic Fh1βKO (red) islets as indicated. Data are presented as area under the curve. ∗p < 0.05 versus 1 mM glucose in CTL mice. #p < 0.05 versus 1 mM glucose in Fh1βKO mice. Data are from indicated number of cells (n) in three islets from one CTL mouse and five islets from two Fh1βKO mice, respectively. All data presented as mean values ± SEM of indicated number of experiments. See also Figure S7.
Figure 4
Figure 4
Hyperglycemia Results in Intracellular Acidification of α Cells (A) Histogram summarizing basal α cell intracellular pH (pHi) measured using the pH indicator seminaphthorhodafluor (SNARF) in α cells in islets from CTL (black) and hyperglycemic Fh1βKO (red) mice. Number of cells indicated below the respective bars in the histogram. ∗p < 0.05 versus CTL. (B) pHi measured in α cells in islets from CTL (black) and hyperglycemic Fh1βKO (red) mice at 3 mM and 20 mM glucose. The traces have been offset to reflect the true difference in fluorescence ratios (F650/550) between CTL and Fh1βKO α cells. Measurements were performed in acutely isolated intact islets. (C) Net effect of 20 mM glucose on pHi (ΔF650/550) in α cells from CTL (black) and nearly normoglycemic (gray) or hyperglycemic (red) Fh1βκO mice. Number of cells (n) indicated above the respective bars in the histogram. ∗p < 0.05 versus CTL; #p < 0.05 versus basal (at 3 mM glucose). (D) Dose-dependent acidification of wild-type α cells in response to ethyl-isopropyl amiloride (EIPA) measured using the pH indicator SNARF. (E) Histogram summarizing the effect of EIPA on α cell pHi in wild-type islets. Number of cells indicated above the respective bars in the histogram. ∗p < 0.05 versus basal (no EIPA). (F) Effect of the non-metabolizable glucose analogue α-methyl-D-glucopyranoside (αMDG) on cytoplasmic Na+ ([Na+]i) in wild-type α cells in the absence (black) and the presence (red) of phlorizin. Glucose (1 mM) was present throughout. [Na+]i was measured using Sodium Green. Fluorescence (F) values have been normalized to that at 1 mM glucose (F0). (G) Effect of αMDG on [Na+]i in the absence (black) and presence (red) of phlorizin in wild-type islets, as indicated. Data in (A) and (B) normalized to Sodium Green fluorescence under basal conditions (1 mM glucose; n = 59 cells) ∗p < 0.05 versus 1 mM glucose; #p < 0.05 versus αMDG in the absence of phlorizin. Data are presented as mean values ±SEM of indicated number of experiments (n). In (B), (D), and (F), representative single-cell traces have been selected for display. See also Figures S2 and S3.
Figure 5
Figure 5
SGLT-Mediated Na+ Uptake Leads to Intracellular Acidification and Reduced ATP Production in the Hyperglycemic α Cell (A) Schematic of ex vivo experiments. Islets are isolated from mouse pancreas and incubated in 11–12 or 20 mM glucose for 48 hr. These concentrations approximate to fed plasma glucose levels before and after hyperglycemia develops in Fh1βKO mice (see Figures S1A and S1B). Figure was made using Servier medical ART. (B) Histogram summarizing basal pHi measured using the pH indicator SNARF in α cells from wild-type islets incubated for 48 hr at 11 mM or 20 mM glucose, or 20 mM glucose plus 50 μM phlorizin. Number of cells (n) indicated below the respective bars in the histogram; ∗p < 0.05 versus 11 mM glucose. #p < 0.05 versus 20 mM glucose. (C) Cytoplasmic ATP/ADP ratio at 1 and 20 mM glucose (indicated above recording) in α cells from wild-type islets cultured for 48 hr at 11 mM or 20 mM glucose, or 20 mM glucose plus 50 μM phlorizin. (D) Histogram summarizing the net effect of glucose on the cytoplasmic ATP/ADP ratio (ΔATP/ADPc) in α cells from wild-type islets cultured at 11 or 20 mM glucose in the absence or presence of 50 μM phlorizin as indicated. ∗p < 0.05 versus 1 mM glucose; #p < 0.05 versus 20 mM glucose in islets cultured at 11 mM glucose. (E) Glucagon secretion measured at 1 or 20 mM glucose in wild-type islets cultured for 48 hr at 12 mM, 20 mM glucose, or 20 mM glucose plus 50 μM phlorizin as indicated. ∗p < 0.05 versus 1 mM glucose in respective group; #p < 0.05 versus 20 mM glucose in islets cultured at 20 mM glucose in the absence of phlorizin (n = 8 experiments using islets from six mice). (F) As in (E) using wild-type islets that were cultured for 48 hr at 12 mM glucose alone (white bars) or in the presence of 10 μM NHE inhibitor EIPA (gray bars) in the absence or presence of 10 μM tolbutamide as indicated. ∗p < 0.05 versus 1 mM glucose in respective group; #p < 0.05 versus 20 mM glucose in islets cultured in the absence of EIPA; #p < 0.05 versus 1 mM glucose EIPA-incubated islets in the absence of tolbutamide (n = 3–8 experiments using islets from five mice). Data are presented as mean values ±SEM of indicated number of experiments (n). Traces in (C) represent average response in all cells. See also Figure S4.
Figure 6
Figure 6
Protein Succination Persists after Restoration of Normoglycemia (A) Glucagon secretion at 1 and 20 mM glucose in acutely isolated islets from CTL and hyperglycemic Fh1βKO mice. ∗p < 0.05 versus 1 mM glucose (n = 9 experiments using islets from four mice of each genotype). (B) As in (A) but after 72 hr of culture at 12 mM glucose. ∗p < 0.05 versus 1 mM glucose (n = 9 experiments for each genotype using islets from four CTL and four Fh1βKO mice). (C) Glucagon content in CTL and Fh1βKO islets either acutely isolated or after 72 hr of culture. ∗p < 0.05 versus CTL. (D) Immunofluorescence for 2SC (green), glucagon (red), insulin (blue), and overlay (yellow) islets from CTL, hyperglycemic βV59M (diabetic), and normoglycemic βV59M mice (treated with glibenclamide). Note strong 2SC labeling of most glucagon-positive cells. Scale bar, 50 μm. (E) IHC for succination (2SC) in islets from non-diabetic (CTL) individuals and patients with type-2 diabetes (T2D). Scale bar, 50 μm. (F) Immunofluorescence for 2SC (green), glucagon (red), DAPI (blue), and overlay (yellow) in islets from patients diagnosed with T2D. Note strong 2SC labeling of most glucagon-positive cells. Scale bar, 50 μm. Data in (E) and (F) are representative of six donors for both the non-diabetic (CTL) and T2D groups. Data are presented as mean values ± SEM of indicated number of experiments (n). See Table S1 for details on the donors.
Figure 7
Figure 7
Protein Succination in Renal Tubular Cells and Cardiomyocytes from Hyperglycemic Fh1βKO Mice (A) IHC for 2SC in CTL (left) and diabetic Fh1βKO (right) hearts showing staining in a subset of cells in diabetic animals but not in the non-diabetic CTLs. Scale bar, 50 μm. (B) As in (A) but showing sections of kidney in CTL (left) and normoglycemic (middle) or hyperglycemic (right) Fh1βKO mice as indicated. (C) Schematic of the proposed relationship between hyperglycemia, impaired ATP production, and protein succination. The increased intracellular fumarate resulting from reduced activity of fumarase leads to protein succination. The sites of action of SGLT inhibitors and tolbutamide are indicated. Sections in (A) and (B) are representative of observations in >10 animals of both genotypes.

References

    1. Adam J., Ramracheya R., Chibalina M.V., Ternette N., Hamilton A., Tarasov A.I., Zhang Q., Rebelato E., Rorsman N.J.G., Martin-Del-Rio R. Fumarate hydratase deletion in pancreatic beta cells leads to progressive diabetes. Cell Rep. 2017;20:3135–3148.
    1. Adam J., Yang M., Bauerschmidt C., Kitagawa M., O’Flaherty L., Maheswaran P., Özkan G., Sahgal N., Baban D., Kato K. A role for cytosolic fumarate hydratase in urea cycle metabolism and renal neoplasia. Cell Rep. 2013;3:1440–1448.
    1. Akhmedov D., Braun M., Mataki C., Park K.S., Pozzan T., Schoonjans K., Rorsman P., Wollheim C.B., Wiederkehr A. Mitochondrial matrix pH controls oxidative phosphorylation and metabolism-secretion coupling in INS-1E clonal beta cells. FASEB J. 2010;24:4613–4626.
    1. Azarias G., Perreten H., Lengacher S., Poburko D., Demaurex N., Magistretti P.J., Chatton J.Y. Glutamate transport decreases mitochondrial pH and modulates oxidative metabolism in astrocytes. J. Neurosci. 2011;31:3550–3559.
    1. Berg J., Hung Y.P., Yellen G. A genetically encoded fluorescent reporter of ATP: ADP ratio. Nat. Methods. 2009;6:161–166.
    1. Bernstein L.H., Everse J. Studies on the mechanism of the malate dehydrogenase reaction. J. Biol. Chem. 1978;253:8702–8707.
    1. Bertero E., Prates Roma L., Ameri P., Maack C. Cardiac effects of SGLT2 inhibitors: the sodium hypothesis. Cardiovasc. Res. 2018;114:12–18.
    1. Blatnik M., Frizzell N., Thorpe S.R., Baynes J.W. Inactivation of glyceraldehyde-3-phosphate dehydrogenase by fumarate in diabetes: formation of S-(2-succinyl)cysteine, a novel chemical modification of protein and possible biomarker of mitochondrial stress. Diabetes. 2008;57:41–49.
    1. Blodgett D.M., Nowosielska A., Afik S., Pechhold S., Cura A.J., Kennedy N.J., Kim S., Kucukural A., Davis R.J., Kent S.C. Novel observations from next-generation RNA sequencing of highly purified human adult and fetal islet cell subsets. Diabetes. 2015;64:3172–3181.
    1. Bonner C., Kerr-Conte J., Gmyr V., Queniat G., Moerman E., Thevenet J., Beaucamps C., Delalleau N., Popescu I., Malaisse W.J. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nat. Med. 2015;21:512–517.
    1. Brereton M.F., Iberl M., Shimomura K., Zhang Q., Adriaenssens A.E., Proks P., Spiliotis I.I., Dace W., Mattis K.K., Ramracheya R. Reversible changes in pancreatic islet structure and function produced by elevated blood glucose. Nat. Commun. 2014;5:4639.
    1. Brown D.A., Perry J.B., Allen M.E., Sabbah H.N., Stauffer B.L., Shaikh S.R., Cleland J.G., Colucci W.S., Butler J., Voors A.A. Expert consensus document: mitochondrial function as a therapeutic target in heart failure. Nat. Rev. Cardiol. 2016;14:238–250.
    1. Casey J.R., Grinstein S., Orlowski J. Sensors and regulators of intracellular pH. Nat. Rev. Mol. Cell Biol. 2010;11:50–61.
    1. Cryer P.E. Managing diabetes: lessons from type 1 diabetes mellitus. Diabet. Med. 1998;15(Suppl 4):S8–S12.
    1. Cryer P.E. Hypoglycaemia: the limiting factor in the glycaemic management of type I and type II diabetes. Diabetologia. 2002;45:937–948.
    1. Cryer P.E. Glycemic goals in diabetes: trade-off between glycemic control and iatrogenic hypoglycemia. Diabetes. 2014;63:2188–2195.
    1. DiGruccio M.R., Mawla A.M., Donaldson C.J., Noguchi G.M., Vaughan J., Cowing-Zitron C., van der Meulen T., Huising M.O. Comprehensive alpha, beta and delta cell transcriptomes reveal that ghrelin selectively activates delta cells and promotes somatostatin release from pancreatic islets. Mol. Metab. 2016;5:449–458.
    1. Dor Y., Brown J., Martinez O.I., Melton D.A. Adult pancreatic β-cells are formed by self-duplication rather than stem-cell differentiation. Nature. 2004;429:41.
    1. Dunning B.E., Foley J.E., Ahren B. Alpha cell function in health and disease: influence of glucagon-like peptide-1. Diabetologia. 2005;48:1700–1713.
    1. Eberhard D., Lammert E. The role of the antioxidant protein DJ-1 in type 2 diabetes mellitus. In: Ariga H., Iguchi-Ariga S.M.M., editors. DJ-1/PARK7 Protein: Parkinson’s Disease, Cancer and Oxidative Stress-Induced Diseases. Springer; 2017. pp. 173–186.
    1. Ferrannini E. Sodium-glucose co-transporters and their inhibition: clinical physiology. Cell Metab. 2017;26:27–38.
    1. Forbes J.M. Mitochondria-power players in kidney function? Trends Endocrinol. Metab. 2016;27:441–442.
    1. Forbes J.M., Cooper M.E. Mechanisms of diabetic complications. Physiol. Rev. 2013;93:137–188.
    1. Frier B.M. The incidence and impact of hypoglycemia in type 1 and type 2 diabetes. International Diabetes Monitor. 2009;21:210–218.
    1. Frizzell N., Lima M., Baynes J.W. Succination of proteins in diabetes. Free Radic. Res. 2011;45:101–109.
    1. Girard C.A., Wunderlich F.T., Shimomura K., Collins S., Kaizik S., Proks P., Abdulkader F., Clark A., Ball V., Zubcevic L. Expression of an activating mutation in the gene encoding the K(ATP) channel subunit Kir6.2 in mouse pancreatic β cells recapitulates neonatal diabetes. J. Clin. Invest. 2009;119:80–90.
    1. Granhall C., Park H.B., Fakhrai-Rad H., Luthman H. High-resolution quantitative trait locus analysis reveals multiple diabetes susceptibility loci mapped to intervals<800 kb in the species-conserved Niddm1i of the GK rat. Genetics. 2006;174:1565–1572.
    1. Hamilton A., Zhang Q., Salehi A., Willems M., Knudsen J.G., Ringgaard A.K., Chapman C.E., Gonzalez-Alvarez A., Surdo N.C., Zaccolo M. Adrenaline stimulates glucagon secretion by Tpc2-dependent Ca2+ mobilization from acidic stores in pancreatic α-cells. Diabetes. 2018;67:1128–1139.
    1. Herrera P.L. Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development. 2000;127:2317–2322.
    1. Jain D., Jain R., Eberhard D., Eglinger J., Bugliani M., Piemonti L., Marchetti P., Lammert E. Age- and diet-dependent requirement of DJ-1 for glucose homeostasis in mice with implications for human type 2 diabetes. J. Mol. Cell Biol. 2012;4:221–230.
    1. Jamison R.A., Stark R., Dong J., Yonemitsu S., Zhang D., Shulman G.I., Kibbey R.G. Hyperglucagonemia precedes a decline in insulin secretion and causes hyperglycemia in chronically glucose-infused rats. Am. J. Physiol. Endocrinol. Metab. 2011;301:E1174–E1183.
    1. Lai J.C.K., Cooper A.J.L. Brain α-ketoglutarate dehydrogenase complex: kinetic properties, regional distribution, and effects of inhibitors. J. Neurochem. 1986;47:1376–1386.
    1. Lopaschuk G.D., Verma S. Empagliflozin's fuel hypothesis: not so soon. Cell Metab. 2016;24:200–202.
    1. Masereel B., Pochet L., Laeckmann D. An overview of inhibitors of Na(+)/H(+) exchanger. Eur. J. Med. Chem. 2003;38:547–554.
    1. Massey V. Studies on fumarase. II. The effects of inorganic anions on fumarase activity. Biochem. J. 1953;53:67–71.
    1. Nagai R., Brock J.W., Blatnik M., Baatz J.E., Bethard J., Walla M.D., Thorpe S.R., Baynes J.W., Frizzell N. Succination of protein thiols during adipocyte maturation: a biomarker of mitochondrial stress. J. Biol. Chem. 2007;282:34219–34228.
    1. Parker H.E., Adriaenssens A., Rogers G., Richards P., Koepsell H., Reimann F., Gribble F.M. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia. 2012;55:2445–2455.
    1. Pollard P.J., Spencer-Dene B., Shukla D., Howarth K., Nye E., El-Bahrawy M., Deheragoda M., Joannou M., McDonald S., Martin A. Targeted inactivation of fh1 causes proliferative renal cyst development and activation of the hypoxia pathway. Cancer Cell. 2007;11:311–319.
    1. Pollard P.J., Wortham N.C., Tomlinson I.P. The TCA cycle and tumorigenesis: the examples of fumarate hydratase and succinate dehydrogenase. Ann. Med. 2003;35:632–639.
    1. R Development Core Team . R Foundation for Statistical Computing; 2016. R: A Language and Environment for Statistical Computing.
    1. Ramracheya R.D., McCulloch L.J., Clark A., Wiggins D., Johannessen H., Olsen M.K., Cai X., Zhao C.M., Chen D., Rorsman P. PYY-dependent restoration of impaired insulin and glucagon secretion in type 2 diabetes following Roux-En-Y gastric bypass surgery. Cell Rep. 2016;15:944–950.
    1. Rorsman P., Ramracheya R., Rorsman N.J., Zhang Q. ATP-regulated potassium channels and voltage-gated calcium channels in pancreatic alpha and beta cells: similar functions but reciprocal effects on secretion. Diabetologia. 2014;57:1749–1761.
    1. Skrivarhaug T., Bangstad H.J., Stene L.C., Sandvik L., Hanssen K.F., Joner G. Long-term mortality in a nationwide cohort of childhood-onset type 1 diabetic patients in Norway. Diabetologia. 2006;49:298–305.
    1. Spegel P., Ekholm E., Tuomi T., Groop L., Mulder H., Filipsson K. Metabolite profiling reveals normal metabolic control in carriers of mutations in the glucokinase gene (MODY2) Diabetes. 2013;62:653–661.
    1. Tarasov A.I., Semplici F., Ravier M.A., Bellomo E.A., Pullen T.J., Gilon P., Sekler I., Rizzuto R., Rutter G.A. The mitochondrial Ca2+ uniporter MCU is essential for glucose-induced ATP increases in pancreatic beta-cells. PLoS One. 2012;7:e39722.
    1. Ternette N., Yang M., Laroyia M., Kitagawa M., O’Flaherty L., Wolhulter K., Igarashi K., Saito K., Kato K., Fischer R. Inhibition of mitochondrial aconitase by succination in fumarate hydratase deficiency. Cell Rep. 2013;3:689–700.
    1. Trube G., Rorsman P., Ohno-Shosaku T. Opposite effects of tolbutamide and diazoxide on the ATP-dependent K+ channel in mouse pancreatic beta-cells. Pflugers Arch. 1986;407:493–499.
    1. Unger R.H., Orci L. Paracrinology of islets and the paracrinopathy of diabetes. Proc. Natl. Acad. Sci. U S A. 2010;107:16009–16012.
    1. Vieira E., Liu Y.J., Gylfe E. Involvement of alpha1 and beta-adrenoceptors in adrenaline stimulation of the glucagon-secreting mouse alpha-cell. Naunyn Schmiedebergs Arch. Pharmacol. 2004;369:179–183.
    1. Walker J.N., Ramracheya R., Zhang Q., Johnson P.R., Braun M., Rorsman P. Regulation of glucagon secretion by glucose: paracrine, intrinsic or both? Diabetes Obes. Metab. 2011;13(Suppl 1):95–105.
    1. Wanner C., Inzucchi S.E., Zinman B. Empagliflozin and progression of kidney disease in type 2 diabetes. N. Engl. J. Med. 2016;375:1801–1802.
    1. Willson V.J.C., Tipton K.F. The effect of pH on the allosteric behaviour of Ox-brain NAD+-dependent isocitrate dehydrogenase. Eur. J. Biochem. 1980;109:411–416.
    1. Wright E.M., Loo D.D., Hirayama B.A. Biology of human sodium glucose transporters. Physiol. Rev. 2011;91:733–794.
    1. Zhang Q., Ramracheya R., Lahmann C., Tarasov A., Bengtsson M., Braha O., Braun M., Brereton M., Collins S., Galvanovskis J. Role of KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in type 2 diabetes. Cell Metab. 2013;18:871–882.
    1. Zinman B., Wanner C., Lachin J.M., Fitchett D., Bluhmki E., Hantel S., Mattheus M., Devins T., Johansen O.E., Woerle H.J. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N. Engl. J. Med. 2015;373:2117–2128.

Source: PubMed

3
Se inscrever