Skip to main content
Top
Published in: Endocrine 2/2024

Open Access 14-11-2023 | Hepatitis C | Original Article

HCV affects KATP channels through GnT-IVa-mediated N-glycosylation of GLUT2 on the surface of pancreatic β-cells leading to impaired insulin secretion

Authors: Ben Niu, Lijing Ma, Lixuan Yao, Yating Zhang, Heng Su

Published in: Endocrine | Issue 2/2024

Login to get access

Abstract

Purpose

To explore the mechanism of insulin secretion dysfunction in pancreatic beta cells induced by N-glycosylation mediated by an infection from the hepatitis C virus (HCV).

Methods

Min6 cell models infected with HCV and stimulated with glucose were constructed. Meanwhile, an HCV-infected animal model and a type 2 diabetes mellitus (T2DM) rat model were constructed. Glucose uptake in the Min6 cells was detected, and insulin secretion was detected by ELISA. Flow cytometry, immunofluorescence staining, Western blotting, RT-qPCR, and lectin blotting were used to detect the expression levels of related proteins and mRNA, as well as the level of N-glycosylation. HE staining was used to observe the pathological changes in the pancreatic tissue, and an oral glucose tolerance test (OGTT) was used to evaluate the glucose tolerance of the rats.

Results

Compared with the NC group, the expression levels of GnT-IVa, GLUT2, galectin-9, and voltage-dependent calcium channel 1.2 (Cav1.2) were significantly downregulated in the HCV-infected group. The ATP-sensitive potassium channel (KATP) component proteins SUR1 and Kir6.2 were significantly upregulated, while intracellular glucose intake and insulin secretion decreased, N-glycosylation levels and ATP levels significantly decreased, and the overexpression of GnT-IVa reversed the effect of the HCV infection. However, treatment with the glycosylation inhibitor kifunensine (KIF) or the KATP channel activator diazine (Dia) reversed the effects of the overexpression of GnT-IVa. In the animal experiments, HE staining revealed serious pathological injuries in the pancreatic tissue of the HCV-infected rats, with decreased glucose tolerance and glycosylation levels, decreased insulin secretion, downregulated expression of GnT-IVa, GLUT2, and Cav1.2, and upregulated expression of SUR1 and Kir6.2. The overexpression treatment of GnT-IVa or the KATP channel antagonist miglinide reversed the effects of HCV.

Conclusion

HCV infection inhibits GLUT2 N-glycosylation on the pancreatic β cell surface by downregulating the expression of GnT-IVa and then activates the KATP pathway, which ultimately leads to disturbances in insulin secretion.
Literature
1.
go back to reference M.L. Shiffman, N.T. Gunn, Impact of hepatitis C virus therapy on metabolism and public health. Liver Int 37, 13–18 (2017)CrossRefPubMed M.L. Shiffman, N.T. Gunn, Impact of hepatitis C virus therapy on metabolism and public health. Liver Int 37, 13–18 (2017)CrossRefPubMed
2.
go back to reference S. Grimbert, P. Valensi, C. Levy-Marchal, G. Perret, J.P. Richardet, C. Raffoux et al. High prevalence of diabetes mellitus in patients with chronic hepatitis C. A case-control study. Gastroenterol Clin Biol 20, 544–548 (1996)PubMed S. Grimbert, P. Valensi, C. Levy-Marchal, G. Perret, J.P. Richardet, C. Raffoux et al. High prevalence of diabetes mellitus in patients with chronic hepatitis C. A case-control study. Gastroenterol Clin Biol 20, 544–548 (1996)PubMed
3.
go back to reference Y. Shintani, H. Fujie, H. Miyoshi, T. Tsutsumi, K. Tsukamoto, S. Kimura et al. Hepatitis C virus infection and diabetes: direct involvement of the virus in the development of insulin resistance. Gastroenterology 126, 840–848 (2004)CrossRefPubMed Y. Shintani, H. Fujie, H. Miyoshi, T. Tsutsumi, K. Tsukamoto, S. Kimura et al. Hepatitis C virus infection and diabetes: direct involvement of the virus in the development of insulin resistance. Gastroenterology 126, 840–848 (2004)CrossRefPubMed
4.
go back to reference L.F. Wang, C.H. Wu, Y. Shan, X.H. Fan, N. Huo, H.Y. Lu et al. Prevalence of abnormal glycometabolism in patients with chronic hepatitis C and related risk factors in China. Chin Med J 124, 183–188 (2011)PubMed L.F. Wang, C.H. Wu, Y. Shan, X.H. Fan, N. Huo, H.Y. Lu et al. Prevalence of abnormal glycometabolism in patients with chronic hepatitis C and related risk factors in China. Chin Med J 124, 183–188 (2011)PubMed
5.
go back to reference J.M. Hui, A. Sud, G.C. Farrell, P. Bandara, K. Byth, J.G. Kench et al. Insulin resistance is associated with chronic hepatitis C virus infection and fibrosis progression [corrected]. Gastroenterology 125, 1695–1704 (2003)CrossRefPubMed J.M. Hui, A. Sud, G.C. Farrell, P. Bandara, K. Byth, J.G. Kench et al. Insulin resistance is associated with chronic hepatitis C virus infection and fibrosis progression [corrected]. Gastroenterology 125, 1695–1704 (2003)CrossRefPubMed
6.
go back to reference K. Maedler, Beta cells in type 2 diabetes - a crucial contribution to pathogenesis. Diabetes Obes Metab 10, 408–420 (2008)CrossRefPubMed K. Maedler, Beta cells in type 2 diabetes - a crucial contribution to pathogenesis. Diabetes Obes Metab 10, 408–420 (2008)CrossRefPubMed
8.
go back to reference D.C. Liemburg-Apers, P.H. Willems, W.J. Koopman, S. Grefte, Interactions between mitochondrial reactive oxygen species and cellular glucose metabolism. Arch Toxicol 89, 1209–1226 (2015)CrossRefPubMedPubMedCentral D.C. Liemburg-Apers, P.H. Willems, W.J. Koopman, S. Grefte, Interactions between mitochondrial reactive oxygen species and cellular glucose metabolism. Arch Toxicol 89, 1209–1226 (2015)CrossRefPubMedPubMedCentral
9.
go back to reference M. Takahashi, Y. Kizuka, K. Ohtsubo, J. Gu, N. Taniguchi, Disease-associated glycans on cell surface proteins. Mol Aspects Med 51, 56–70 (2016)CrossRefPubMed M. Takahashi, Y. Kizuka, K. Ohtsubo, J. Gu, N. Taniguchi, Disease-associated glycans on cell surface proteins. Mol Aspects Med 51, 56–70 (2016)CrossRefPubMed
10.
go back to reference K. Ohtsubo, S. Takamatsu, C. Gao, H. Korekane, T.M. Kurosawa, N. Taniguchi, N-Glycosylation modulates the membrane sub-domain distribution and activity of glucose transporter 2 in pancreatic beta cells. Biochem Biophys Res Commun 434, 346–351 (2013)CrossRefPubMed K. Ohtsubo, S. Takamatsu, C. Gao, H. Korekane, T.M. Kurosawa, N. Taniguchi, N-Glycosylation modulates the membrane sub-domain distribution and activity of glucose transporter 2 in pancreatic beta cells. Biochem Biophys Res Commun 434, 346–351 (2013)CrossRefPubMed
11.
go back to reference F.M. Ashcroft, D.E. Harrison, S.J. Ashcroft, Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature 312, 446–448 (1984)CrossRefPubMed F.M. Ashcroft, D.E. Harrison, S.J. Ashcroft, Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature 312, 446–448 (1984)CrossRefPubMed
12.
go back to reference M. Hiriart, M. Velasco, C. Larque, C.M. Diaz-Garcia, Metabolic syndrome and ionic channels in pancreatic beta cells. Vitam Horm 95, 87–114 (2014)CrossRefPubMed M. Hiriart, M. Velasco, C. Larque, C.M. Diaz-Garcia, Metabolic syndrome and ionic channels in pancreatic beta cells. Vitam Horm 95, 87–114 (2014)CrossRefPubMed
13.
go back to reference D. Kasai, T. Adachi, L. Deng, M. Nagano-Fujii, K. Sada, M. Ikeda et al. HCV replication suppresses cellular glucose uptake through down-regulation of cell surface expression of glucose transporters. J Hepatol 50, 883–894 (2009)CrossRefPubMed D. Kasai, T. Adachi, L. Deng, M. Nagano-Fujii, K. Sada, M. Ikeda et al. HCV replication suppresses cellular glucose uptake through down-regulation of cell surface expression of glucose transporters. J Hepatol 50, 883–894 (2009)CrossRefPubMed
14.
go back to reference S. Trivedi, S. Murthy, H. Sharma, A.S. Hartlage, A. Kumar, S.V. Gadi et al. Viral persistence, liver disease, and host response in a hepatitis C-like virus rat model. Hepatology 68, 435–448 (2018)CrossRefPubMed S. Trivedi, S. Murthy, H. Sharma, A.S. Hartlage, A. Kumar, S.V. Gadi et al. Viral persistence, liver disease, and host response in a hepatitis C-like virus rat model. Hepatology 68, 435–448 (2018)CrossRefPubMed
15.
go back to reference C. Sumi, A. Okamoto, H. Tanaka, M. Kusunoki, T. Shoji, T. Uba et al. Suppression of mitochondrial oxygen metabolism mediated by the transcription factor HIF-1 alleviates propofol-induced cell toxicity. Sci Rep 8, 8987 (2018)CrossRefPubMedPubMedCentral C. Sumi, A. Okamoto, H. Tanaka, M. Kusunoki, T. Shoji, T. Uba et al. Suppression of mitochondrial oxygen metabolism mediated by the transcription factor HIF-1 alleviates propofol-induced cell toxicity. Sci Rep 8, 8987 (2018)CrossRefPubMedPubMedCentral
16.
go back to reference K. Date, A. Satoh, K. Iida, H. Ogawa, Pancreatic alpha-Amylase Controls Glucose Assimilation by Duodenal Retrieval through N-Glycan-specific Binding, Endocytosis, and Degradation. J Biol Chem 290, 17439–17450 (2015)CrossRefPubMedPubMedCentral K. Date, A. Satoh, K. Iida, H. Ogawa, Pancreatic alpha-Amylase Controls Glucose Assimilation by Duodenal Retrieval through N-Glycan-specific Binding, Endocytosis, and Degradation. J Biol Chem 290, 17439–17450 (2015)CrossRefPubMedPubMedCentral
17.
go back to reference K. Schaefer, N.E. Webb, M. Pang, J.E. Hernandez-Davies, K.P. Lee, P. Gonzalez et al. Galectin-9 binds to O-glycans on protein disulfide isomerase. Glycobiology 27, 878–887 (2017)CrossRefPubMedPubMedCentral K. Schaefer, N.E. Webb, M. Pang, J.E. Hernandez-Davies, K.P. Lee, P. Gonzalez et al. Galectin-9 binds to O-glycans on protein disulfide isomerase. Glycobiology 27, 878–887 (2017)CrossRefPubMedPubMedCentral
18.
go back to reference J.C. Koster, M.A. Permutt, C.G. Nichols, Diabetes and insulin secretion: the ATP-sensitive K+ channel (K ATP) connection. Diabetes 54, 3065–3072 (2005)CrossRefPubMed J.C. Koster, M.A. Permutt, C.G. Nichols, Diabetes and insulin secretion: the ATP-sensitive K+ channel (K ATP) connection. Diabetes 54, 3065–3072 (2005)CrossRefPubMed
19.
go back to reference J.P. Messina, I. Humphreys, A. Flaxman, A. Brown, G.S. Cooke, O.G. Pybus et al. Global distribution and prevalence of hepatitis C virus genotypes. Hepatology 61, 77–87 (2015)CrossRefPubMed J.P. Messina, I. Humphreys, A. Flaxman, A. Brown, G.S. Cooke, O.G. Pybus et al. Global distribution and prevalence of hepatitis C virus genotypes. Hepatology 61, 77–87 (2015)CrossRefPubMed
20.
go back to reference S.H. Mehta, F.L. Brancati, M.S. Sulkowski, S.A. Strathdee, M. Szklo, D.L. Thomas, Prevalence of type 2 diabetes mellitus among persons with hepatitis C virus infection in the United States. Ann Intern Med 133, 592–599 (2000)CrossRefPubMed S.H. Mehta, F.L. Brancati, M.S. Sulkowski, S.A. Strathdee, M. Szklo, D.L. Thomas, Prevalence of type 2 diabetes mellitus among persons with hepatitis C virus infection in the United States. Ann Intern Med 133, 592–599 (2000)CrossRefPubMed
21.
go back to reference H. Ma, G.P. You, X.P. Zhang, X.J. Yang, H.D. Lu, Y.L. Huang et al. A novel role of globular adiponectin in treatment with HFD/STZ induced T2DM combined with NAFLD rats. TheScientificWorldJournal 2014, 230835 (2014)CrossRefPubMedPubMedCentral H. Ma, G.P. You, X.P. Zhang, X.J. Yang, H.D. Lu, Y.L. Huang et al. A novel role of globular adiponectin in treatment with HFD/STZ induced T2DM combined with NAFLD rats. TheScientificWorldJournal 2014, 230835 (2014)CrossRefPubMedPubMedCentral
22.
go back to reference M.J. Hossain, M.D. Kendig, M.E. Letton, M.J. Morris, R. Arnold, Peripheral Neuropathy Phenotyping in Rat Models of Type 2 Diabetes Mellitus: Evaluating Uptake of the Neurodiab Guidelines and Identifying Future Directions. Diabetes Metab J 46, 198–221 (2022)CrossRefPubMedPubMedCentral M.J. Hossain, M.D. Kendig, M.E. Letton, M.J. Morris, R. Arnold, Peripheral Neuropathy Phenotyping in Rat Models of Type 2 Diabetes Mellitus: Evaluating Uptake of the Neurodiab Guidelines and Identifying Future Directions. Diabetes Metab J 46, 198–221 (2022)CrossRefPubMedPubMedCentral
23.
go back to reference X. Wang, Q. Li, X. Han, M. Gong, Z. Yu, B. Xu, Electroacupuncture Alleviates Diabetic Peripheral Neuropathy by Regulating Glycolipid-Related GLO/AGEs/RAGE Axis. Front Endocrinol 12, 655591 (2021)CrossRef X. Wang, Q. Li, X. Han, M. Gong, Z. Yu, B. Xu, Electroacupuncture Alleviates Diabetic Peripheral Neuropathy by Regulating Glycolipid-Related GLO/AGEs/RAGE Axis. Front Endocrinol 12, 655591 (2021)CrossRef
24.
go back to reference A. Lecube, C. Hernandez, J. Genesca, J.I. Esteban, R. Jardi, R. Simo, High prevalence of glucose abnormalities in patients with hepatitis C virus infection: a multivariate analysis considering the liver injury. Diabetes Care 27, 1171–1175 (2004)CrossRefPubMed A. Lecube, C. Hernandez, J. Genesca, J.I. Esteban, R. Jardi, R. Simo, High prevalence of glucose abnormalities in patients with hepatitis C virus infection: a multivariate analysis considering the liver injury. Diabetes Care 27, 1171–1175 (2004)CrossRefPubMed
25.
go back to reference M.L. Macheda, S. Rogers, J.D. Best, Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol 202, 654–662 (2005)CrossRefPubMed M.L. Macheda, S. Rogers, J.D. Best, Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol 202, 654–662 (2005)CrossRefPubMed
26.
go back to reference N. Ban, Y. Yamada, Y. Someya, K. Miyawaki, Y. Ihara, M. Hosokawa et al. Hepatocyte nuclear factor-1alpha recruits the transcriptional co-activator p300 on the GLUT2 gene promoter. Diabetes 51, 1409–1418 (2002)CrossRefPubMed N. Ban, Y. Yamada, Y. Someya, K. Miyawaki, Y. Ihara, M. Hosokawa et al. Hepatocyte nuclear factor-1alpha recruits the transcriptional co-activator p300 on the GLUT2 gene promoter. Diabetes 51, 1409–1418 (2002)CrossRefPubMed
27.
go back to reference C. Matsui, I. Shoji, S. Kaneda, I.R. Sianipar, L. Deng, H. Hotta, Hepatitis C virus infection suppresses GLUT2 gene expression via downregulation of hepatocyte nuclear factor 1alpha. J Virol 86, 12903–12911 (2012)CrossRefPubMedPubMedCentral C. Matsui, I. Shoji, S. Kaneda, I.R. Sianipar, L. Deng, H. Hotta, Hepatitis C virus infection suppresses GLUT2 gene expression via downregulation of hepatocyte nuclear factor 1alpha. J Virol 86, 12903–12911 (2012)CrossRefPubMedPubMedCentral
28.
go back to reference K. Ohtsubo, S. Takamatsu, M.T. Minowa, A. Yoshida, M. Takeuchi, J.D. Marth, Dietary and genetic control of glucose transporter 2 glycosylation promotes insulin secretion in suppressing diabetes. Cell 123, 1307–1321 (2005)CrossRefPubMed K. Ohtsubo, S. Takamatsu, M.T. Minowa, A. Yoshida, M. Takeuchi, J.D. Marth, Dietary and genetic control of glucose transporter 2 glycosylation promotes insulin secretion in suppressing diabetes. Cell 123, 1307–1321 (2005)CrossRefPubMed
29.
go back to reference M. Kusunoki, M. Hayashi, T. Shoji, T. Uba, H. Tanaka, C. Sumi et al. Propofol inhibits stromatoxin-1-sensitive voltage-dependent K(+) channels in pancreatic beta-cells and enhances insulin secretion. PeerJ 7, e8157 (2019)CrossRefPubMedPubMedCentral M. Kusunoki, M. Hayashi, T. Shoji, T. Uba, H. Tanaka, C. Sumi et al. Propofol inhibits stromatoxin-1-sensitive voltage-dependent K(+) channels in pancreatic beta-cells and enhances insulin secretion. PeerJ 7, e8157 (2019)CrossRefPubMedPubMedCentral
30.
go back to reference C.M. Driggers, S.L. Shyng, Production and purification of ATP-sensitive potassium channel particles for cryo-electron microscopy. Methods Enzymol 653, 121–150 (2021)CrossRefPubMedPubMedCentral C.M. Driggers, S.L. Shyng, Production and purification of ATP-sensitive potassium channel particles for cryo-electron microscopy. Methods Enzymol 653, 121–150 (2021)CrossRefPubMedPubMedCentral
31.
go back to reference E.B. Pratt, Q. Zhou, J.W. Gay, S.L. Shyng, Engineered interaction between SUR1 and Kir6.2 that enhances ATP sensitivity in KATP channels. J Gen Physiol 140, 175–187 (2012)CrossRefPubMedPubMedCentral E.B. Pratt, Q. Zhou, J.W. Gay, S.L. Shyng, Engineered interaction between SUR1 and Kir6.2 that enhances ATP sensitivity in KATP channels. J Gen Physiol 140, 175–187 (2012)CrossRefPubMedPubMedCentral
32.
go back to reference H. Ishihara, T. Asano, K. Tsukuda, H. Katagiri, K. Inukai, M. Anai et al. Pancreatic beta cell line MIN6 exhibits characteristics of glucose metabolism and glucose-stimulated insulin secretion similar to those of normal islets. Diabetologia 36, 1139–1145 (1993)CrossRefPubMed H. Ishihara, T. Asano, K. Tsukuda, H. Katagiri, K. Inukai, M. Anai et al. Pancreatic beta cell line MIN6 exhibits characteristics of glucose metabolism and glucose-stimulated insulin secretion similar to those of normal islets. Diabetologia 36, 1139–1145 (1993)CrossRefPubMed
Metadata
Title
HCV affects KATP channels through GnT-IVa-mediated N-glycosylation of GLUT2 on the surface of pancreatic β-cells leading to impaired insulin secretion
Authors
Ben Niu
Lijing Ma
Lixuan Yao
Yating Zhang
Heng Su
Publication date
14-11-2023
Publisher
Springer US
Published in
Endocrine / Issue 2/2024
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-023-03589-z

Other articles of this Issue 2/2024

Endocrine 2/2024 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine