Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. May 15, 2024; 15(5): 814-817
Published online May 15, 2024. doi: 10.4239/wjd.v15.i5.814
Glucokinase regulatory protein rs780094 polymorphism is associated with type 2 diabetes mellitus, dyslipidemia, non-alcoholic fatty liver disease, and nephropathy
Ashraf Al Madhoun, Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15400, Kuwait
ORCID number: Ashraf Al Madhoun (0000-0001-8593-3878).
Author contributions: Al Madhoun A designed the overall concept, review of literature, writing, and editing the manuscript.
Supported by the Kuwait Foundation for the Advancement of Sciences (KFAS) and Dasman Diabetes Institute, No. RACB-2021-007.
Conflict-of-interest statement: The authors declare that they have no conflict of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ashraf Al Madhoun, PhD, Academic Editor, Senior Scientist, Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Jassim AlBahar Street, Dasman 15400, Kuwait. ashraf.madhoun@dasmaninstitute.org
Received: December 4, 2023
Peer-review started: December 4, 2023
First decision: January 23, 2024
Revised: January 31, 2024
Accepted: March 11, 2024
Article in press: March 11, 2024
Published online: May 15, 2024

Abstract

In this editorial, we comment on the article by Liu et al published in the recent issue of the World Journal of Diabetes (Relationship between GCKR gene rs780094 polymorphism and type 2 diabetes with albuminuria). Type 2 diabetes mellitus (T2DM) is a chronic disorder characterized by dysregulated glucose homeostasis. The persistent elevated blood glucose level in T2DM significantly increases the risk of developing severe complications, including cardiovascular disease, re-tinopathy, neuropathy, and nephropathy. T2DM arises from a complex interplay between genetic, epigenetic, and environmental factors. Global genomic studies have identified numerous genetic variations associated with an increased risk of T2DM. Specifically, variations within the glucokinase regulatory protein (GCKR) gene have been linked to heightened susceptibility to T2DM and its associated complications. The clinical trial by Liu et al further elucidates the role of the GCKR rs780094 polymorphism in T2DM and nephropathy development. Their findings demonstrate that individuals carrying the CT or TT genotype at the GCKR rs780094 locus are at a higher risk of developing T2DM with albuminuria compared to those with the CC genotype. These findings highlight the importance of genetic testing and risk assessment in T2DM to develop effective preventive strategies and personalized treatment plans.

Key Words: Glucokinase regulatory protein, rs780094, Type 2 diabetes mellitus, Dyslipidemia, Non-alcoholic fatty liver disease, Nephropathy

Core Tip: Genome-wide association studies have identified the glucokinase regulatory protein rs780094 variant as a risk factor for type 2 diabetes mellitus and its associated dyslipidemia, non-alcoholic fatty liver disease, and nephropathy. The precise mechanism of action remains elusive. Clinical evidence suggests that the variant impairs pancreatic β-cell function and disrupts hepatic triglyceride and glucose metabolism across various ethnicities.



INTRODUCTION

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by impaired glucose homeostasis, essential for maintaining stable blood glucose levels[1]. This disruption arises from a combination of underlying patho-physiological mechanisms, such as insulin resistance (IR) and diminished glucose-stimulated insulin secretion (GSIS)[2]. Consequently, the interplay between IR and impaired GSIS leads to elevated fasting plasma glucose concentrations and increased hepatic glucose production, culminating in chronic hyperglycemia, a hallmark of T2DM[3]. In this editorial, we comment on the article by Liu et al[4] published in the recent issue of the World Journal of Diabetes.

Glycolysis is a fundamental metabolic pathway that converts glucose into pyruvate, which then enters the citric acid cycle for energy production[5]. Alterations in this multi-step process can dysregulate glucose homeostasis and may lead to T2DM. Glucokinase (GK) catalyzes the phosphorylation of glucose to glucose-6-phosphate which is a pivotal step in glycolysis. This irreversible reaction traps glucose within the cell and commits it to the glycolytic pathway. GK is crucial in pancreatic β-cells and the liver, where it acts as a key regulator of GSIS and glucose uptake and storage, respectively[6]. GK exhibits multi-level regulation, encompassing transcriptional and post-transcriptional mechanisms[7]. Fasting induces nuclear sequestration of GK in an inactive complex with the inhibitory protein GK regulatory protein (GCKR)[8]. Contrarywise, postprandial glucose influx promotes dissociation of the GK/GCKR complex, triggering GK to translocation to the cytoplasm for activation[8]. Thus, the GK–GCKR complex functions as a metabolic “switch”, initiating energy storage and release pathways in response to fasting/feeding status. T2DM is associated with multiple perturbations in GK regulation, including reduced gene expression, impaired compartmental shuttling mediated by GCKR, and/or dysregulation of other metabolic and hormonal pathways that normally influence GK activity[9,10], as comprehensively reviewed in a study[7].

Genome-wide association studies have identified associations between genetic variants in the GCKR gene and T2DM. These variants influence protein function through various molecular mechanisms, ultimately leading to aberrant GK activation during normoglycemia or hypoglycemia. The most extensively studied variant is rs780094, located within intron 16 of the GCKR gene. This locus disrupts binding sites for several hepatocyte-specific transcription factors, including members of the hepatic nuclear factor and Forkhead/winged-helix families, potentially regulating GCKR gene transcription. However, these regulatory interactions require further functional validation.

The rs780094 polymorphism is implicated in the risk of T2DM and dyslipidemia across various populations. In a European cohort study, researchers examined 386731 common SNPs in 2931 participants, evenly split between diabetic patients and healthy controls. They discovered a significant association between the GCKR variant rs780094 and serum triglycerides[11]. In subsequent research, the authors expanded their scope to 12 independent cohorts, totaling over 45000 individuals from diverse non-European ethnic backgrounds[11]. This more extensive study indicated that the rs780094 variant exerts opposing effects on fasting plasma triglyceride and glucose levels and correlates with CRP concentrations[12].

A cohort study of Danish subjects found that the presence of the minor A-allele in the rs780094 was linked to elevated fasting serum triacylglycerol levels, compromised insulin secretion during both fasting and OGTT, and lower HOMA-IR scores[13]. Qi et al[14] reported similar findings in a cohort of 3210 elderly Han Chinese individuals (aged between 50 and 70 years) recruited from the “Study on Nutrition and Health of Aging Population in China”. This clinical investigation revealed that the A-allele, in rs780094, exhibited a marginal association with a decreased incidence of T2DM. However, it was significantly associated with a lower risk of impaired fasting glucose, lower fasting glucose levels, and enhanced β-cell function as measured by HOMA-β[14,15]. Notably, the A-allele was also associated with elevated fasting triacylglycerol levels, but not with HOMA of insulin sensitivity[14,15]. Comparable results were reported in Chinese adults and adolescents[16] and in Japanese populations[17].

The association of the rs780094 variant is limited to the incidence of metabolic diseases and does not extend to coronary heart disease[18]. A recent study investigated the link between rs780094 and lipid accumulation, which is crucial in liver disease, particularly non-alcoholic fatty liver disease (NAFLD)[19]. Silva et al[19] conducted a metabolomics data analysis using samples from the Finnish METSIM clinical trial to comprehensively identify metabolites associated with the rs780094 variant. The findings indicated that the rs780094 minor A-allele is inversely associated with glycerophospholipids, sphingolipids, gamma-glutamylthreonine, and taurocholenate sulfates. In contrast, the same allele was positively associated with retinol, indolelactate, and N-acetyltryptophan, as well as with 12 glycerolipids and 19 glycerophospholipids[19].

CONCLUSION

The pleiotropy of the GCKR rs780094 polymorphism is significantly associated with metabolic diseases, including T2DM, its associated dyslipidemia, NAFLD, and nephropathy. Although the rs780094 variant is believed to alter GCKR gene expression, its precise mechanism of action remains elusive. Clinical evidence suggests that this variant impairs pancreatic β-cell function and disrupts hepatic triglyceride and glucose metabolism across various populations. Therefore, considering the interplay of the GCKR rs780094 polymorphism with environmental and lifestyle factors represents a promising approach to precision medicine, offering an innovative strategy for tailoring disease prevention and treatment.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: Kuwait

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Wang K, China S-Editor: Chen YL L-Editor: A P-Editor: Chen YX

References
1.  ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Cusi K, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA; American Diabetes Association. Addendum. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Care in Diabetes-2023. Diabetes Care 2023;46(Suppl. 1):S49-S67. Diabetes Care. 2023;46:1718-1720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
2.  Deshpande AD, Harris-Hayes M, Schootman M. Epidemiology of diabetes and diabetes-related complications. Phys Ther. 2008;88:1254-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 896]  [Cited by in F6Publishing: 853]  [Article Influence: 53.3]  [Reference Citation Analysis (0)]
3.  Park SY, Gautier JF, Chon S. Assessment of Insulin Secretion and Insulin Resistance in Human. Diabetes Metab J. 2021;45:641-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
4.  Liu YY, Wan Q. Relationship between GCKR gene rs780094 polymorphism and type 2 diabetes with albuminuria. World J Diabetes. 2023;14:1803-1812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Chandel NS. Glycolysis. Cold Spring Harb Perspect Biol. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 63]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
6.  Al-Hasani H, Tschöp MH, Cushman SW. Two birds with one stone: novel glucokinase activator stimulates glucose-induced pancreatic insulin secretion and augments hepatic glucose metabolism. Mol Interv. 2003;3:367-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
7.  Agius L. Glucokinase and molecular aspects of liver glycogen metabolism. Biochem J. 2008;414:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 258]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
8.  van Schaftingen E, Veiga-da-Cunha M, Niculescu L. The regulatory protein of glucokinase. Biochem Soc Trans. 1997;25:136-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 55]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
9.  Shin JS, Torres TP, Catlin RL, Donahue EP, Shiota M. A defect in glucose-induced dissociation of glucokinase from the regulatory protein in Zucker diabetic fatty rats in the early stage of diabetes. Am J Physiol Regul Integr Comp Physiol. 2007;292:R1381-R1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
10.  Payne VA, Arden C, Lange AJ, Agius L. Contributions of glucokinase and phosphofructokinase-2/fructose bisphosphatase-2 to the elevated glycolysis in hepatocytes from Zucker fa/fa rats. Am J Physiol Regul Integr Comp Physiol. 2007;293:R618-R625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
11.  Diabetes Genetics Initiative of Broad Institute of Harvard and MIT; Lund University, and Novartis Institutes of BioMedical Research, Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, Daly MJ, Hughes TE, Groop L, Altshuler D, Almgren P, Florez JC, Meyer J, Ardlie K, Bengtsson Boström K, Isomaa B, Lettre G, Lindblad U, Lyon HN, Melander O, Newton-Cheh C, Nilsson P, Orho-Melander M, Råstam L, Speliotes EK, Taskinen MR, Tuomi T, Guiducci C, Berglund A, Carlson J, Gianniny L, Hackett R, Hall L, Holmkvist J, Laurila E, Sjögren M, Sterner M, Surti A, Svensson M, Tewhey R, Blumenstiel B, Parkin M, Defelice M, Barry R, Brodeur W, Camarata J, Chia N, Fava M, Gibbons J, Handsaker B, Healy C, Nguyen K, Gates C, Sougnez C, Gage D, Nizzari M, Gabriel SB, Chirn GW, Ma Q, Parikh H, Richardson D, Ricke D, Purcell S. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science. 2007;316:1331-1336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2124]  [Cited by in F6Publishing: 2085]  [Article Influence: 122.6]  [Reference Citation Analysis (0)]
12.  Orho-Melander M, Melander O, Guiducci C, Perez-Martinez P, Corella D, Roos C, Tewhey R, Rieder MJ, Hall J, Abecasis G, Tai ES, Welch C, Arnett DK, Lyssenko V, Lindholm E, Saxena R, de Bakker PI, Burtt N, Voight BF, Hirschhorn JN, Tucker KL, Hedner T, Tuomi T, Isomaa B, Eriksson KF, Taskinen MR, Wahlstrand B, Hughes TE, Parnell LD, Lai CQ, Berglund G, Peltonen L, Vartiainen E, Jousilahti P, Havulinna AS, Salomaa V, Nilsson P, Groop L, Altshuler D, Ordovas JM, Kathiresan S. Common missense variant in the glucokinase regulatory protein gene is associated with increased plasma triglyceride and C-reactive protein but lower fasting glucose concentrations. Diabetes. 2008;57:3112-3121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 222]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
13.  Sparsø T, Andersen G, Nielsen T, Burgdorf KS, Gjesing AP, Nielsen AL, Albrechtsen A, Rasmussen SS, Jørgensen T, Borch-Johnsen K, Sandbaek A, Lauritzen T, Madsbad S, Hansen T, Pedersen O. The GCKR rs780094 polymorphism is associated with elevated fasting serum triacylglycerol, reduced fasting and OGTT-related insulinaemia, and reduced risk of type 2 diabetes. Diabetologia. 2008;51:70-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 121]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
14.  Qi Q, Wu Y, Li H, Loos RJ, Hu FB, Sun L, Lu L, Pan A, Liu C, Wu H, Chen L, Yu Z, Lin X. Association of GCKR rs780094, alone or in combination with GCK rs1799884, with type 2 diabetes and related traits in a Han Chinese population. Diabetologia. 2009;52:834-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 49]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
15.  Ling Y, Li X, Gu Q, Chen H, Lu D, Gao X. Associations of common polymorphisms in GCKR with type 2 diabetes and related traits in a Han Chinese population: a case-control study. BMC Med Genet. 2011;12:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
16.  Tam CH, Ma RC, So WY, Wang Y, Lam VK, Germer S, Martin M, Chan JC, Ng MC. Interaction effect of genetic polymorphisms in glucokinase (GCK) and glucokinase regulatory protein (GCKR) on metabolic traits in healthy Chinese adults and adolescents. Diabetes. 2009;58:765-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
17.  Onuma H, Tabara Y, Kawamoto R, Shimizu I, Kawamura R, Takata Y, Nishida W, Ohashi J, Miki T, Kohara K, Makino H, Osawa H. The GCKR rs780094 polymorphism is associated with susceptibility of type 2 diabetes, reduced fasting plasma glucose levels, increased triglycerides levels and lower HOMA-IR in Japanese population. J Hum Genet. 2010;55:600-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
18.  Bi M, Kao WH, Boerwinkle E, Hoogeveen RC, Rasmussen-Torvik LJ, Astor BC, North KE, Coresh J, Köttgen A. Association of rs780094 in GCKR with metabolic traits and incident diabetes and cardiovascular disease: the ARIC Study. PLoS One. 2010;5:e11690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
19.  Fernandes Silva L, Vangipurapu J, Kuulasmaa T, Laakso M. An intronic variant in the GCKR gene is associated with multiple lipids. Sci Rep. 2019;9:10240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]