Next Article in Journal
Pro-Angiogenic Bioactive Molecules in Vascular Morphogenesis: Integrating Endothelial Cell Dynamics
Previous Article in Journal
Identification and Association of CYP2R1, CYP27B1, and GC Gene Polymorphisms with Vitamin D Deficiency in Apparently Healthy Population and in Silico Analysis of the Binding Pocket of Vitamin D3
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Significant Association Between Glucokinase Regulatory Protein Variants and Genetic and Metabolic Diseases

1
Division of Cardiology, Department of Internal Medicine and the Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(10), 850; https://doi.org/10.3390/cimb47100850
Submission received: 24 July 2025 / Revised: 4 October 2025 / Accepted: 5 October 2025 / Published: 15 October 2025

Abstract

As next-generation sequencing develops, there are significant associations between glucokinase regulatory protein (GCKR) variants and many diseases, especially metabolic diseases. However, there is a lack of solid descriptions and summaries of how GCKR variants lead to diseases and a lack of successful translations of drugs targeting this molecular variant. We searched literature datasets, mainly including PubMed and Web of Science, with “GCKR” or “GKRP”, “Variants”, “Hypertriglyceridemia”, “NAFLD”, and “Metabolic diseases” as the search terms. Our review firstly introduces the biological function of the GCKR gene and its encoding protein GKRP and then describes the GCKR variants in different diseases, such as hypertriglyceridemia and NAFLD, revealing that GCKR/GKPR is strongly associated with metabolic diseases. GKPR might be a potential target for T2D and other metabolic diseases. One drug for interfering with the GCK-GKRP complex has been developed and has shown its effectiveness in preclinical studies, with some possible side effects. More and more different-structured drugs should be developed to improve side effects, and more clinical trials should be carried out to determine the best intervention window and timing points to improve prognosis. Taken together, these insights show that GCKR/GKRP is significantly associated with many metabolic diseases via its complex metabolism system and is a potential target in many metabolic diseases.

1. Brief Introduction to Glucokinase Regulatory Protein

Glucokinase regulatory protein (GKRP), encoded by the GCKR gene, is biologically and genetically involved in key metabolic pathways [1]. Located in the nucleus of hepatocytes, GKRP binds to and inhibits glucokinase (GCK), thereby helping regulate blood glucose levels by controlling glucose uptake and storage (Figure 1). GCK acts as a critical regulator of glucose disposal and storage in both the liver and pancreatic beta-cells. It responds to rising blood glucose concentrations by initiating a cascade that leads to insulin secretion from beta-cells and subsequent hepatic glucose uptake and storage. As an inhibitor of GCK—an essential enzyme that phosphorylates glucose to glucose-6-phosphate (the first step of glycolysis)—GKRP plays a central regulatory role [2]. Notably, GKRP is expressed exclusively in the liver, despite the abundance of GCK in the pancreas, suggesting that GKRP modulates glucose and lipid metabolism specifically in the liver rather than the pancreas [3,4]. Under fasting or hypoglycemic conditions, GKRP binds GCK and sequesters it in the nucleus, limiting the conversion of glucose to glucose-6-phosphate and thereby increasing blood glucose levels. This nuclear sequestration of GCK by GKRP suppresses hepatic glycolysis, making more glucose available for other organs. In contrast, under high-glucose conditions, GKRP’s affinity for GCK decreases, releasing GCK into the cytoplasm, where it promotes glycogen synthesis and de novo lipogenesis [5,6,7]. The interaction between GKRP and GCK is further modulated by metabolic intermediates: fructose-6-phosphate (F6P) enhances their binding and suppresses glycolysis, while fructose-1-phosphate (F1P) inhibits this interaction [2]. Enhancing GKRP function may lower blood glucose levels, offering a potential therapeutic strategy in diabetes [8]. Moreover, GKRP helps stabilize GCK, as GCK degradation is observed in GKRP-knockout models [9,10]. Overall, GKRP critically regulates glucose metabolism by controlling the binding, localization, and activity of GCK.
With the completion of the Human Genome Project and advances in next-generation sequencing, numerous studies have identified significant associations between GCKR variants and metabolic diseases, including type 2 diabetes, non-alcoholic fatty liver disease (NAFLD), and familial hypertriglyceridemia [11,12,13]. This review summarizes the functional impacts of GCKR genetic variants on metabolism and discusses potential therapeutic strategies targeting GKRP.

2. Glucokinase Regulatory Variants and Hypertriglyceridemia

Hypertriglyceridemia, a common form of hyperlipidemia, is typically defined as a fasting serum triglyceride level of 150 mg/dL (1.7 mmol/L) or higher, according to the American Heart Association (AHA) 2011 guidelines [14,15,16]. It represents the most prevalent type of dyslipidemia in the general population. Data from the NHANES 2003–2006 survey indicate that approximately 53% of U.S. adults have dyslipidemia, among whom 30% exhibit elevated serum triglycerides (>1.7 mmol/L), while 27% have high serum low-density lipoprotein (LDL) cholesterol levels [17]. Excess lipid deposition in various organs can lead to conditions such as fatty liver disease and pancreatitis [17]. Moreover, hyperlipidemia is recognized as a major risk factor for cardiovascular diseases, including atherosclerosis, myocardial infarction, and heart failure [18,19]. Hypertriglyceridemia arises from factors such as a sedentary lifestyle, poor dietary habits, and genetic variants. Numerous genome-wide association studies (GWASs) and whole-exome sequencing (WES) analyses have identified several genes such as Apolipoprotein A5 (APOA5), Lipoprotein lipase (LPL), Apolipoprotein C2 (APOC2), Familial lipase maturation factor 1 (LMF1), and Glycosylphosphatidylinositol-Anchored High-Density (GPIHBP1), variants of which lead to hypertriglyceridemia [20,21,22], while variants of Apolipoprotein B, (APOB), Low-density lipoprotein receptor (LDLR), and Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) lead to hypercholesteremia [23,24,25,26].
Among these, GCKR variants, initially recognized for their role in glucose metabolism, have attracted increasing attention. Although GKRP was first characterized as a glycolysis regulator, recent genomic studies have strongly linked its variants to triglyceride levels. Stable isotope studies suggest that hepatic fat accumulation may enhance the synthesis of very-low-density lipoprotein (VLDL). Loss-of-function variants in GCKR may reduce its binding affinity for glucose and promote hepatic fat accumulation, thereby increasing VLDL secretion [27,28]. Fat accumulation in the liver also stimulates the production of apolipoprotein B (APOB), a key component of VLDL particles [29]. After secretion, VLDL is progressively converted to intermediate-density lipoprotein (IDL) and then to LDL. Cholesteryl ester transfer protein (CETP) facilitates the exchange of triglycerides and cholesteryl esters among VLDL, LDL, and HDL particles. This process can result in elevated LDL and reduced HDL levels, ultimately contributing to hyperlipidemia (Figure 2).
Hyperlipidemia, especially hypertriglyceridemia, is strongly associated with GCKR variants according to current studies. The non-synonymous variant rs1260326 (p. Pro446Leu) and the intronic SNP rs780094 are two common variants [30,31].
The common variant rs1260326 has been detected in many GWASs. The fine mapping of the GCKR locus revealed that SNP rs1260326 exhibited the strongest correlation with triglyceride concentrations and showed a strong linkage disequilibrium [31,32]. Furthermore, several GWASs on diabetes and hyperlipidemia have also found this variant. According to the ClinVar database, the global minor allele frequency (MAF) of rs1260326 is almost 0.3, indicating that roughly one in three individuals carries this variant. Rees reported that rs1260326 significantly downregulated the expression of GKRP and its interaction with glucokinase in the absence of any intervention, similar to when fructose-1-phosphate (F1P) and fructose-6-phosphate (F6P) were administered [33]. In addition, no difference was observed in the direct affinity between F1P and F6P. Zelent et al. found that glucose, fructose 1-phosphate, and glucokinase activators decreased the affinity of GK for GKRP and increased the binding cooperativity. These findings reflected the disruption of the GK–GKRP complex, while TF-based biophysical analysis revealed that GKRP-P446L might impair the GCK-GKRP complex and nuclear storage [34]. This may explain the inverse change in serum glucose and triglyceride in the fasting state. Despite the increasing number of GCKR variants discovered via NGS, rs1260326 continues to play a pivotal role, prompting additional research into the functions and phenotypes of GCKR variants.
Compared with rs1260326, rs780094 is less researched due to its position on an intron. This variant correlates with the expression of GCKR and the occurrence of hyperlipidemia, with a minor allele frequency (MAF) ranging from 0.2 to 0.4 according to various databases. Moreover, the SNP rs780094 is associated with higher triglyceride, apoB, and CRP levels but not LDL or HDL cholesterol levels [31]. The loci located in a liver-specific enhancer and haploid deficiency contribute to associated traits. In addition, Forkhead box A2 (FOXA2) combines with the enhancer and elevates H3K27Ac levels to promote GCKR transcriptional activity. In the rs780094 variant, the T allele replaces the C allele, which may result in decreased binding to FOXA2 and GCKR transcriptional activitiy [35].
Detection and functional analysis of the above two variants have provided mechanistic insights into the pathways affected by the GCKR variations. In addition to the above common variants, other rare variants have also been detected in genome studies. Johansen identified rare GCKR variants in 438 individuals with hypertriglyceridemia compared with the control cohort [32]. Rees et al. also extended the atlas of GCKR variants and presented a model for interpreting the clinical significance of rare genetic variants in common diseases in 2012 [36]. They identified 10 novel rare coding variants by sequencing the exome of GCKR in 800 coronary atherosclerosis patients from the ClinSeq cohort [36]. The following functional analysis of the 18 rare non-synonymous GCKR variants was conducted by biological experiments and bioinformatic prediction, but the lack of consistency between the two analyses emphasized the low predictive value of rare GCKR variants and the complex heritability of lipid traits [33]. Western blot analysis was performed to study different expression levels of GKPR, and homogenous time-resolved fluorescence and microscale thermophoresis were used to compare the affinity of recombinant WT and variant GKRP for GCK, F1P, and F6P. Subsequently, the Ribbon model of the F1P-bound form of human F1P was run. The inconsistent results between bioinformatic predictions and functional assays for rare variants underscore the challenges in their clinical interpretation. This suggests that the pathogenicity of rare GCKR variants cannot be reliably predicted in silico and requires functional validation. Consequently, the clinical utility of testing rare GCKR variants in hypertriglyceridemia remains limited until more robust variant-specific evidence is available. This stands in contrast to the well-established association of common variants like rs1260326.
In a recent study, Ford et al. used adenoviral vectors for human or mouse GCKR. P446L was transfected in hepatocytes and diet-challenged P446L mice, revealing that the diet-challenged P446L mice developed several traits found to correspond to the rs1260326 locus on chromosome 2, including raised blood cholesterol, lower blood glucose, and low-er liver glucokinase and GKRP, but not raised blood triglycerides [37]. This discrepancy with human association studies might be attributed to differences in murine and human lipid metabolism and the specific dietary challenge employed. The different genotypic milieu between human and mice and the instability of the model system should not be ignored. In addition, the different GKRP perturbation in the model system should be considered; AAV infection is acute and short-term, while the GCKR variant causes injury from birth. Maybe an earlier infection time point could be tried, or GCKR point mutation models might be a better choice. Further research is needed to reconcile these findings.
According to many studies and case reports, GCKR variants do not cosegregate with triglycerides in hypertriglyceridemic pedigrees [33,38]. It is theorized that hereditary disease may be caused by the interaction between different gene mutations or other risk factors, which may provide evidence for Knudson’s two-hit hypothesis [39]. In addition to different gene mutations, the different loci of the same gene may contribute to disease occurrence as well. A study in 2016 reported that a patient carried three distinct GCKR mutation sites and suffered from severe hyperlipidemia following her pregnancy [38]. These findings suggest that primary familiar hypertriglyceridemia may arise from a complex genetic basis, as they are often caused by the interaction between several different gene variants or environmental factors instead of an individual gene variant [40,41].

3. Glucokinase Regulatory Protein and Non-Alcoholic Fatty Liver Disease

Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver condition worldwide, with a prevalence of approximately 25% in the adult population [42]. It is recognized as a hepatic manifestation of metabolic syndrome and is frequently associated with obesity, insulin resistance, and dyslipidemia. NAFLD encompasses a spectrum ranging from simple hepatic steatosis to inflammation and hepatocyte ballooning, primarily resulting from non-secondary hepatic fat accumulation [43]. In recent years, genome-wide association studies (GWASs), whole-exome sequencing (WES), candidate gene analyses, and family-based studies have indicated a significant genetic component underlying NAFLD. Family studies suggest that genetic factors account for at least 38% of the variability in hepatic fat content and NAFLD susceptibility [44,45]. Several gene loci have been implicated, including the GCKR variants rs1260326 (P446L) and rs780094 [46,47].
Notably, GCKR mRNA expression is significantly reduced in patients with severe hepatic steatosis compared to those with mild steatosis [48], highlighting the need for further research to identify pathogenic GCKR variants and elucidate their role in NAFLD pathogenesis. Both in vitro and in vivo studies suggest that the rs1260326 polymorphism impairs the inhibitory function of GKRP toward GCK, leading to enhanced hepatic glucose uptake, accelerated de novo lipogenesis, and ultimately increased hepatic fat accumulation [3,41,49,50]. Meanwhile, rs780094 is located within a liver-specific intragenic enhancer that modulates GCKR expression in a haplotype-dependent manner [35]. Strong linkage disequilibrium between rs1260326 and rs780094 suggests that these variants may collectively increase NAFLD risk [31]. A large-scale meta-analysis involving 16,927 participants (5115 cases and 11,812 controls) for rs780094 and 11,233 participants (2238 cases and 8995 controls) for rs1260326 further confirmed their significant association with NAFLD, with odds ratios of 1.20 (95% CI: 1.11–1.29) for rs780094 and 1.32 (95% CI: 1.22–1.42) for rs1260326 [30].

4. Glucokinase Regulatory Protein and Metabolic Syndrome

Although NAFLD is considered a component of metabolic syndrome, GCKR variants have also been significantly associated with a broader range of metabolic abnormalities. These variants influence an extensive array of metabolites involved in carbohydrate, fatty acid, amino acid, purine, and lipid metabolism (Figure 1 and Figure 2) [51,52,53,54,55], as summarized in the review by Brouwers et al. [28]. A meta-analysis of 40,000 subjects showed that homozygous carriers of the rs780094 variant had 1.9% lower plasma glucose levels but 13% higher triglyceride concentrations compared to non-carriers [31]. Interestingly, GCKR variants exhibit a distinct relationship with type 2 diabetes (T2D) relative to other metabolic disorders. For instance, some GCKR variants that are protective against T2D have been positively associated with elevated branched-chain amino acids such as isoleucine and leucine, which may conversely increase T2D risk [56,57,58]. Additionally, genetic studies have linked GCKR variants to higher levels of C-reactive protein (CRP) and coagulation factors such as protein C and factor VII [59,60,61,62]. The relationship between GCKR variants and CRP may be partly mediated by NAFLD. CRP, an acute-phase inflammatory protein, is produced in response to IL-6 [63]. Elevated IL-6 and CRP levels are common in obesity, and although direct evidence linking GCKR variants to obesity is limited, this pathway warrants further investigation [64]. One report suggested that CRP levels are more closely associated with hepatic fat content than with obesity itself, implying that NAFLD may contribute to increased CRP levels [65].

5. Glucokinase Regulatory Protein and Diabetes Mellitus

Diabetes mellitus is a complex disease influenced by multiple genetic and environmental factors, complicating the identification of its underlying causes and the development of effective therapies. To date, genome-wide association studies (GWASs) and other genetic approaches have identified several susceptibility loci, including peroxisome proliferator-activated receptor gamma (PPARG), glucagon-like peptide 1 receptor (GLP1R), and transcription factor 7-like 2 (TCF7L2) [66,67,68]. Some of these discoveries have already translated into therapeutic targets; for example, PPARG is the target of insulin-sensitizing thiazolidinediones, and small-molecule GLP1R agonists have gained widespread clinical use [69,70]. Pathogenic variants in GCK are known causes of maturity-onset diabetes of the young (MODY) [71,72]. Functional studies indicate that these variants reduce glucose affinity, impairing beta-cell insulin secretion and hepatic glycogen storage [73,74]. These findings support the potential utility of GCK activators in diabetes treatment, though their use is limited by a significant risk of hypoglycemia. To overcome this limitation, Lloyd et al. (2013) developed a small molecule designed to disrupt the GCK–GKRP interaction [75]. Since GKRP is expressed exclusively in the liver, this compound selectively activates hepatic GCK without affecting pancreatic beta-cells, thereby lowering fasting glucose levels with a reduced risk of hypoglycemia [76]. Using a cell-free high-throughput screening approach, the team identified a molecule that binds GKRP and promotes GCK translocation from the nucleus to the cytoplasm in primary rat hepatocytes [75]. The compound effectively reduced serum glucose levels in hyperglycemic rats but not in normoglycemic controls [75]. Subsequent studies in diet-induced obese C57BL/6 mice, as well as ob/ob and db/db models, yielded consistent results [77]. While these findings highlight the therapeutic potential of targeting the GCK–GKRP complex, potential adverse effects must be carefully considered. Studies suggest that loss-of-function GCKR variants may disrupt lipid homeostasis over time, indicating that pharmacological inhibition of GKRP could perturb lipid metabolism [10,78]. The indications and contraindications of such a drug therefore require thorough evaluation. In addition, the relationship between GCKR variants and cardiovascular outcomes remains unclear. Although rare loss-of-function GCKR variants do not consistently cosegregate with hypertriglyceridemia, their influence on lipid levels should not be overlooked [33]. For instance, while one study reported that the rs780094 variant may protect against cardiovascular disease, other large-scale studies have reached conflicting conclusions [79,80,81]. In one follow-up analysis, GCKR polymorphism was associated with an increased cardiovascular risk, though a significant association was observed only for ischemic cardiomyopathy after adjustment for lipid levels and T2D status—a finding that may limit its reliability [81].
A notable paradox arises between the goal of pharmacologically inducing GKRP loss-of-function to lower blood glucose and the adverse metabolic profile associated with genetic GCKR loss-of-function [82]. This discrepancy may stem from the chronic nature of genetic deficiency versus the acute pharmacological effect, as well as the complex interplay of metabolic pathways involved [83]. A lower dosage and extended treatment regimen may help mitigate side effects, though large-scale clinical trials will be essential to define the therapeutic window and optimal dosing. In summary, further clinical studies are needed to evaluate the efficacy, safety, and overall risk–benefit profile of GKRP-targeting glucose-lowering agents. Genetic screening may also play a future role in personalizing treatment and identifying patients most likely to respond safely to this approach.

6. Limitations and Future Studies

Although numerous studies have established the association between GCKR variants and metabolic diseases, confirming both the pathogenicity of these variants and the therapeutic potential of targeting GKRP, translational progress into clinical practice remains limited. The development of small molecules that disrupt the GCK–GKRP complex represents a promising therapeutic strategy, potentially offering a novel approach to T2D management without the risk of systemic hypoglycemia. Such targeted agents echo the success of other genetics-driven therapies, such as PCSK9 inhibitors for lipid management [84,85]. However, the potential side effects of GKRP modulation—particularly on lipid homeostasis—must be carefully addressed through further preclinical and clinical studies. Notably, emerging multi-target agents such as GLP-1R/GCGR dual agonists are already under investigation for metabolic and liver diseases [86], highlighting the growing interest in pathway-based therapies. Given the complex role of GKRP within the metabolic system, minimizing side effects—particularly on lipid metabolism—poses a significant challenge. Future research should focus on identifying optimal therapeutic windows, including appropriate dosing and treatment timing, through well-designed clinical trials. Additionally, artificial intelligence could aid in designing more precise drug structures, while alternative therapeutic platforms such as antisense oligodeoxynotides (ASOs) or adeno-associated virus (AAV)-based approaches may offer new avenues. Concurrently, further genetic studies on GCKR variants across different diseases will help clarify the clinical significance of GKRP and strengthen the evidence base for its therapeutic targeting.

7. Conclusions

The GCKR gene and its encoded protein, GKRP, play essential roles in multiple metabolic pathways, including glucose, lipid, and uric acid metabolism [87,88]. Both common and rare GCKR variants have been linked to hypertriglyceridemia, NAFLD, and type 2 diabetes, underscoring their broad impact on human metabolic health. Among these, the rs1260326 variant stands out due to its strong genetic association and functional relevance across a spectrum of metabolic diseases. With advances in next-generation sequencing and increasing numbers of family-based and population genetic studies, more GCKR variants are likely to be identified, enabling deeper functional characterization and clearer mechanistic insights. With continued research, GKRP-targeted interventions may eventually find a place in clinical practice, offering new options for patients with metabolic disorders.

8. Methods in the Search Strategy

The literature datasets mainly included PubMed and Web of Science, and the search terms included “GCKR” or “GKRP”, “Variants”, “Hypertriglyceridemia”, “NAFLD”, and “Metabolic diseases”. The inclusion criteria were that (1) the article comprised original research or a case report; (2) there was a comprehensive meta-analysis; (3) it described and investigated significant associations between GCKR variants and metabolic diseases. The exclusion criteria were that the article (1) showed a lack of solid variants and sequencing evidence; (2) was published before the 1980s.

Author Contributions

P.C. and H.W. made contributions to the design of the review. Y.C. and X.S. selected the data for this report. B.Y., Y.S. and K.X. analyzed and interpreted the data. K.X. drafted the manuscript. All the authors have read and approved the final manuscript, and all the authors have agreed both to be personally accountable for their own contributions and to ensure that questions related to the accuracy or integrity of any part of this case report, even ones in which the author was not personally involved, are appropriately investigated and resolved and the resolution documented in the literature. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by NSFC projects (Nos.82100526 for Bo Yu and Nos.82100510 for Peng Chen) and Hubei Provincial Natural Science Foundation of China (No. 2025AFB589 for Bo Yu).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Van Schaftingen, E. A protein from rat liver confers to glucokinase the property of being antagonistically regulated by fructose 6-phosphate and fructose 1-phosphate. Eur. J. Biochem. 1989, 179, 179–184. [Google Scholar] [CrossRef]
  2. Veiga-da-Cunha, M.; Van Schaftingen, E. Identification of fructose 6-phosphate- and fructose 1-phosphate-binding residues in the regulatory protein of glucokinase. J. Biol. Chem. 2002, 277, 8466–8473. [Google Scholar] [CrossRef]
  3. Beer, N.L.; Tribble, N.D.; McCulloch, L.J.; Roos, C.; Johnson, P.R.; Orho-Melander, M.; Gloyn, A.L. The P446L variant in GCKR associated with fasting plasma glucose and triglyceride levels exerts its effect through increased glucokinase activity in liver. Hum. Mol. Genet. 2009, 18, 4081–4088. [Google Scholar] [CrossRef]
  4. Nica, A.C.; Ongen, H.; Irminger, J.C.; Bosco, D.; Berney, T.; Antonarakis, S.E.; Halban, P.A.; Dermitzakis, E.T. Cell-type, allelic, and genetic signatures in the human pancreatic beta cell transcriptome. Genome Res. 2013, 23, 1554–1562. [Google Scholar] [CrossRef]
  5. Agius, L. Hormonal and Metabolite Regulation of Hepatic Glucokinase. Annu. Rev. Nutr. 2016, 36, 389–415. [Google Scholar] [CrossRef]
  6. Detheux, M.; Vandercammen, A.; Van Schaftingen, E. Effectors of the regulatory protein acting on liver glucokinase: A kinetic investigation. Eur. J. Biochem. 1991, 200, 553–561. [Google Scholar] [CrossRef]
  7. Vandercammen, A.; Detheux, M.; Van Schaftingen, E. Binding of sorbitol 6-phosphate and of fructose 1-phosphate to the regulatory protein of liver glucokinase. Biochem. J. 1992, 286 Pt 1, 253–256. [Google Scholar] [CrossRef]
  8. van Schaftingen, E.; Vandercammen, A.; Detheux, M.; Davies, D.R. The regulatory protein of liver glucokinase. Adv. Enzyme Regul. 1992, 32, 133–148. [Google Scholar] [CrossRef] [PubMed]
  9. Grimsby, J.; Coffey, J.W.; Dvorozniak, M.T.; Magram, J.; Li, G.; Matschinsky, F.M.; Shiota, C.; Kaur, S.; Magnuson, M.A.; Grippo, J.F. Characterization of glucokinase regulatory protein-deficient mice. J. Biol. Chem. 2000, 275, 7826–7831. [Google Scholar] [CrossRef] [PubMed]
  10. Farrelly, D.; Brown, K.S.; Tieman, A.; Ren, J.; Lira, S.A.; Hagan, D.; Gregg, R.; Mookhtiar, K.A.; Hariharan, N. Mice mutant for glucokinase regulatory protein exhibit decreased liver glucokinase: A sequestration mechanism in metabolic regulation. Proc. Natl. Acad. Sci. USA 1999, 96, 14511–14516. [Google Scholar] [CrossRef] [PubMed]
  11. Allison, M.E. Fatty liver. Hosp. Med. 2004, 65, 609–612. [Google Scholar] [CrossRef] [PubMed]
  12. Ioannou, G.N. Epidemiology and risk-stratification of NAFLD-associated HCC. J. Hepatol. 2021, 75, 1476–1484. [Google Scholar] [CrossRef]
  13. Ghodsian, N.; Abner, E.; Emdin, C.A.; Gobeil, É.; Taba, N.; Haas, M.E.; Perrot, N.; Manikpurage, H.D.; Gagnon, É.; Bourgault, J.; et al. Electronic health record-based genome-wide meta-analysis provides insights on the genetic architecture of non-alcoholic fatty liver disease. Cell Rep. Med. 2021, 2, 100437. [Google Scholar] [CrossRef]
  14. Miller, M.; Stone, N.J.; Ballantyne, C.; Bittner, V.; Criqui, M.H.; Ginsberg, H.N.; Goldberg, A.C.; Howard, W.J.; Jacobson, M.S.; Kris-Etherton, P.M.; et al. Triglycerides and cardiovascular disease: A scientific statement from the American Heart Association. Circulation 2011, 123, 2292–2333. [Google Scholar] [CrossRef]
  15. Simha, V. Management of hypertriglyceridemia. BMJ 2020, 371, m3109. [Google Scholar] [CrossRef]
  16. Karr, S. Epidemiology and management of hyperlipidemia. Am. J. Manag. Care 2017, 23 (Suppl. 9), S139–S148. [Google Scholar]
  17. Tóth, P.P.; Potter, D.; Ming, E.E. Prevalence of lipid abnormalities in the United States: The National Health and Nutrition Examination Survey 2003–2006. J. Clin. Lipidol. 2012, 6, 325–330. [Google Scholar] [CrossRef]
  18. Tietge, U.J. Hyperlipidemia and cardiovascular disease: Inflammation, dyslipidemia, and atherosclerosis. Curr. Opin. Lipidol. 2014, 25, 94–95. [Google Scholar] [CrossRef] [PubMed]
  19. Nordestgaard, B.G.; Varbo, A. Triglycerides and cardiovascular disease. Lancet 2014, 384, 626–635. [Google Scholar] [CrossRef] [PubMed]
  20. Chyzhyk, V.; Brown, A.S. Familial chylomicronemia syndrome: A rare but devastating autosomal recessive disorder characterized by refractory hypertriglyceridemia and recurrent pancreatitis. Trends Cardiovasc. Med. 2020, 30, 80–85. [Google Scholar]
  21. Ariza, M.J.; Coca-Prieto, I.; Rioja, J.; Muñiz-Grijalvo, O.; Zambón-Rados, D.; Blanco-Echevarría, A.; Arrobas-Velilla, T.; Delgado-Lista, J.; León-Jiménez, D.; Casañas-Martínez, M.; et al. Pathogenicity assessment of genetic variants identified in patients with severe hypertriglyceridemia: Novel cases of familial chylomicronemia syndrome from the Dyslipidemia Registry of the Spanish Atherosclerosis Society. Genet. Med. 2025, 27, 101365. [Google Scholar]
  22. Dron, J.S.; Hegele, R.A. Genetics of Hypertriglyceridemia. Front Endocrinol 2020, 11, 455. [Google Scholar]
  23. Sanghera, D.K.; Hopkins, R.; Malone-Perez, M.W.; Bejar, C.; Tan, C.; Mussa, H.; Whitby, P.; Fowler, B.; Rao, C.V.; Fung, K.A.; et al. Targeted sequencing of candidate genes of dyslipidemia in Punjabi Sikhs: Population-specific rare variants in GCKR promote ectopic fat deposition. PLoS ONE 2019, 14, e0211661. [Google Scholar] [CrossRef]
  24. Alnouri, F.; Athar, M.; Al-Allaf, F.A.; Abduljaleel, Z.; Taher, M.M.; Bouazzaoui, A.; Al Ammari, D.; Karrar, H.; Albabtain, M. Novel combined variants of LDLR and LDLRAP1 genes causing severe familial hypercholesterolemia. Atherosclerosis 2018, 277, 425–433. [Google Scholar]
  25. Shaik, N.A.; Al-Qahtani, F.; Nasser, K.; Jamil, K.; Alrayes, N.M.; Elango, R.; Awan, Z.A.; Banaganapalli, B. Molecular insights into the coding region mutations of low-density lipoprotein receptor adaptor protein 1 (LDLRAP1) linked to familial hypercholesterolemia. J. Gene Med. 2020, 22, e3176. [Google Scholar]
  26. Chemello, K.; García-Nafría, J.; Gallo, A.; Martín, C.; Lambert, G.; Blom, D. Lipoprotein metabolism in familial hypercholesterolemia. J. Lipid Res. 2021, 62, 100062. [Google Scholar]
  27. Adiels, M.; Taskinen, M.R.; Packard, C.; Caslake, M.J.; Soro-Paavonen, A.; Westerbacka, J.; Vehkavaara, S.; Häkkinen, A.; Olofsson, S.O.; Yki-Järvinen, H.; et al. Overproduction of large VLDL particles is driven by increased liver fat content in man. Diabetologia 2006, 49, 755–765. [Google Scholar] [CrossRef]
  28. Brouwers, M.; Jacobs, C.; Bast, A.; Stehouwer, C.D.A.; Schaper, N.C. Modulation of Glucokinase Regulatory Protein: A Double-Edged Sword? Trends Mol. Med. 2015, 21, 583–594. [Google Scholar] [CrossRef]
  29. Ruscica, M.; Ferri, N.; Macchi, C.; Meroni, M.; Lanti, C.; Ricci, C.; Maggioni, M.; Fracanzani, A.L.; Badiali, S.; Fargion, S.; et al. Liver fat accumulation is associated with circulating PCSK9. Ann. Med. 2016, 48, 384–391. [Google Scholar]
  30. Li, J.; Zhao, Y.; Zhang, H.; Hua, W.; Jiao, W.; Du, X.; Rui, J.; Li, S.; Teng, H.; Shi, B.; et al. Contribution of Rs780094 and Rs1260326 Polymorphisms in GCKR Gene to Non-alcoholic Fatty Liver Disease: A Meta-Analysis Involving 26,552 Participants. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 1696–1708. [Google Scholar] [CrossRef]
  31. Orho-Melander, M.; Melander, O.; Guiducci, C.; Perez-Martinez, P.; Corella, D.; Roos, C.; Tewhey, R.; Rieder, M.J.; Hall, J.; Abecasis, G.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  32. Johansen, C.T.; Wang, J.; Lanktree, M.B.; Cao, H.; McIntyre, A.D.; Ban, M.R.; Martins, R.A.; Kennedy, B.A.; Hassell, R.G.; Visser, M.E.; et al. Excess of rare variants in genes identified by genome-wide association study of hypertriglyceridemia. Nat. Genet. 2010, 42, 684–687. [Google Scholar] [CrossRef] [PubMed]
  33. Rees, M.G.; Raimondo, A.; Wang, J.; Ban, M.R.; Davis, M.I.; Barrett, A.; Ranft, J.; Jagdhuhn, D.; Waterstradt, R.; Baltrusch, S.; et al. Inheritance of rare functional GCKR variants and their contribution to triglyceride levels in families. Hum. Mol. Genet. 2014, 23, 5570–5578. [Google Scholar] [CrossRef] [PubMed]
  34. Zelent, B.; Raimondo, A.; Barrett, A.; Buettger, C.W.; Chen, P.; Gloyn, A.L.; Matschinsky, F.M. Analysis of the co-operative interaction between the allosterically regulated proteins GK and GKRP using tryptophan fluorescence. Biochem. J. 2014, 459, 551–564. [Google Scholar] [CrossRef]
  35. López Rodríguez, M.; Kaminska, D.; Lappalainen, K.; Pihlajamäki, J.; Kaikkonen, M.U.; Laakso, M. Identification and characterization of a FOXA2-regulated transcriptional enhancer at a type 2 diabetes intronic locus that controls GCKR expression in liver cells. Genome Med. 2017, 9, 63. [Google Scholar] [CrossRef]
  36. Rees, M.G.; Ng, D.; Ruppert, S.; Turner, C.; Beer, N.L.; Swift, A.J.; Morken, M.A.; Below, J.E.; Blech, I.; Mullikin, J.C.; et al. Correlation of rare coding variants in the gene encoding human glucokinase regulatory protein with phenotypic, cellular, and kinetic outcomes. J. Clin. Investig. 2012, 122, 205–217. [Google Scholar] [CrossRef]
  37. Ford, B.E.; Chachra, S.S.; Rodgers, K.; Moonira, T.; Al-Oanzi, Z.H.; Anstee, Q.M.; Reeves, H.L.; Schattenberg, J.M.; Fairclough, R.J.; Smith, D.M.; et al. The GCKR-P446L gene variant predisposes to raised blood cholesterol and lower blood glucose in the P446L mouse-a model for GCKR rs1260326. Mol. Metab. 2023, 72, 101722. [Google Scholar] [CrossRef]
  38. Shetty, S.; Xing, C.; Garg, A. Type 1 Hyperlipoproteinemia Due to Compound Heterozygous Rare Variants in GCKR. J. Clin. Endocrinol. Metab. 2016, 101, 3884–3887. [Google Scholar] [CrossRef] [PubMed]
  39. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  40. Lewis, G.F.; Xiao, C.; Hegele, R.A. Hypertriglyceridemia in the genomic era: A new paradigm. Endocr. Rev. 2015, 36, 131–147. [Google Scholar] [CrossRef]
  41. Brouwers, M.C.; van Greevenbroek, M.M.; Stehouwer, C.D.; de Graaf, J.; Stalenhoef, A.F. The genetics of familial combined hyperlipidaemia. Nat. Rev. Endocrinol. 2012, 8, 352–362. [Google Scholar] [CrossRef]
  42. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef]
  43. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [Google Scholar] [CrossRef]
  44. Schwimmer, J.B.; Celedon, M.A.; Lavine, J.E.; Salem, R.; Campbell, N.; Schork, N.J.; Shiehmorteza, M.; Yokoo, T.; Chavez, A.; Middleton, M.S.; et al. Heritability of nonalcoholic fatty liver disease. Gastroenterology 2009, 136, 1585–1592. [Google Scholar] [CrossRef]
  45. Willner, I.R.; Waters, B.; Patil, S.R.; Reuben, A.; Morelli, J.; Riely, C.A. Ninety patients with nonalcoholic steatohepatitis: Insulin resistance, familial tendency, and severity of disease. Am. J. Gastroenterol. 2001, 96, 2957–2961. [Google Scholar] [CrossRef]
  46. Speliotes, E.K.; Yerges-Armstrong, L.M.; Wu, J.; Hernaez, R.; Kim, L.J.; Palmer, C.D.; Gudnason, V.; Eiriksdottir, G.; Garcia, M.E.; Launer, L.J.; et al. Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS Genet. 2011, 7, e1001324. [Google Scholar] [CrossRef] [PubMed]
  47. Santoro, N.; Zhang, C.K.; Zhao, H.; Pakstis, A.J.; Kim, G.; Kursawe, R.; Dykas, D.J.; Bale, A.E.; Giannini, C.; Pierpont, B.; et al. Variant in the glucokinase regulatory protein (GCKR) gene is associated with fatty liver in obese children and adolescents. Hepatology 2012, 55, 781–789. [Google Scholar] [CrossRef]
  48. Bechmann, L.P.; Gastaldelli, A.; Vetter, D.; Patman, G.L.; Pascoe, L.; Hannivoort, R.A.; Lee, U.E.; Fiel, I.; Muñoz, U.; Ciociaro, D.; et al. Glucokinase links Krüppel-like factor 6 to the regulation of hepatic insulin sensitivity in nonalcoholic fatty liver disease. Hepatology 2012, 55, 1083–1093. [Google Scholar] [CrossRef]
  49. Rees, M.G.; Wincovitch, S.; Schultz, J.; Waterstradt, R.; Beer, N.L.; Baltrusch, S.; Collins, F.S.; Gloyn, A.L. Cellular characterisation of the GCKR P446L variant associated with type 2 diabetes risk. Diabetologia 2012, 55, 114–122. [Google Scholar] [CrossRef]
  50. Simons, N.; Dekker, J.M.; van Greevenbroek, M.M.; Nijpels, G.; t Hart, L.M.; van der Kallen, C.J.; Schalkwijk, C.G.; Schaper, N.C.; Stehouwer, C.D.; Brouwers, M.C. A Common Gene Variant in Glucokinase Regulatory Protein Interacts with Glucose Metabolism on Diabetic Dyslipidemia: The Combined CODAM and Hoorn Studies. Diabetes Care 2016, 39, 1811–1817. [Google Scholar] [CrossRef]
  51. Bi, M.; Kao, W.H.; Boerwinkle, E.; Hoogeveen, R.C.; Rasmussen-Torvik, L.J.; Astor, B.C.; North, K.E.; 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. [Google Scholar] [CrossRef]
  52. Qi, Q.; Wu, Y.; Li, H.; Loos, R.J.; Hu, F.B.; Sun, L.; Lu, L.; Pan, A.; Liu, C.; Wu, H.; et al. 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. [Google Scholar] [CrossRef]
  53. Vaxillaire, M.; Cavalcanti-Proença, C.; Dechaume, A.; Tichet, J.; Marre, M.; Balkau, B.; Froguel, P. The common P446L polymorphism in GCKR inversely modulates fasting glucose and triglyceride levels and reduces type 2 diabetes risk in the DESIR prospective general French population. Diabetes 2008, 57, 2253–2257. [Google Scholar] [CrossRef]
  54. Kozian, D.H.; Barthel, A.; Cousin, E.; Brunnhöfer, R.; Anderka, O.; März, W.; Böhm, B.; Winkelmann, B.; Bornstein, S.R.; Schmoll, D. Glucokinase-activating GCKR polymorphisms increase plasma levels of triglycerides and free fatty acids, but do not elevate cardiovascular risk in the Ludwigshafen Risk and Cardiovascular Health Study. Horm. Metab. Res. 2010, 42, 502–506. [Google Scholar] [CrossRef] [PubMed]
  55. Mahendran, Y.; Vangipurapu, J.; Cederberg, H.; Stancáková, A.; Pihlajamäki, J.; Soininen, P.; Kangas, A.J.; Paananen, J.; Civelek, M.; Saleem, N.K.; et al. Association of ketone body levels with hyperglycemia and type 2 diabetes in 9398 Finnish men. Diabetes 2013, 62, 3618–3626. [Google Scholar] [CrossRef]
  56. Stancáková, A.; Civelek, M.; Saleem, N.K.; Soininen, P.; Kangas, A.J.; Cederberg, H.; Paananen, J.; Pihlajamäki, J.; Bonnycastle, L.L.; Morken, M.A.; et al. Hyperglycemia and a common variant of GCKR are associated with the levels of eight amino acids in 9369 Finnish men. Diabetes 2012, 61, 1895–1902. [Google Scholar] [CrossRef] [PubMed]
  57. Chambers, J.C.; Zhang, W.; Sehmi, J.; Li, X.; Wass, M.N.; Van der Harst, P.; Holm, H.; Sanna, S.; Kavousi, M.; Baumeister, S.E.; et al. Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma. Nat. Genet. 2011, 43, 1131–1138. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, T.J.; Larson, M.G.; Vasan, R.S.; Cheng, S.; Rhee, E.P.; McCabe, E.; Lewis, G.D.; Fox, C.S.; Jacques, P.F.; Fernandez, C.; et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 2011, 17, 448–453. [Google Scholar] [CrossRef] [PubMed]
  59. Ridker, P.M.; Pare, G.; Parker, A.; Zee, R.Y.; Danik, J.S.; Buring, J.E.; Kwiatkowski, D.; Cook, N.R.; Miletich, J.P.; Chasman, D.I. Loci related to metabolic-syndrome pathways including LEPR, HNF1A, IL6R, and GCKR associate with plasma C-reactive protein: The Women’s Genome Health Study. Am. J. Hum. Genet. 2008, 82, 1185–1192. [Google Scholar] [CrossRef] [PubMed]
  60. Dehghan, A.; Dupuis, J.; Barbalic, M.; Bis, J.C.; Eiriksdottir, G.; Lu, C.; Pellikka, N.; Wallaschofski, H.; Kettunen, J.; Henneman, P.; et al. Meta-analysis of genome-wide association studies in >80,000 subjects identifies multiple loci for C-reactive protein levels. Circulation 2011, 123, 731–738. [Google Scholar] [CrossRef]
  61. Taylor, K.C.; Lange, L.A.; Zabaneh, D.; Lange, E.; Keating, B.J.; Tang, W.; Smith, N.L.; Delaney, J.A.; Kumari, M.; Hingorani, A.; et al. A gene-centric association scan for Coagulation Factor VII levels in European and African Americans: The Candidate Gene Association Resource (CARe) Consortium. Hum. Mol. Genet. 2011, 20, 3525–3534. [Google Scholar] [CrossRef]
  62. Smith, N.L.; Chen, M.H.; Dehghan, A.; Strachan, D.P.; Basu, S.; Soranzo, N.; Hayward, C.; Rudan, I.; Sabater-Lleal, M.; Bis, J.C.; et al. Novel associations of multiple genetic loci with plasma levels of factor VII, factor VIII, and von Willebrand factor: The CHARGE (Cohorts for Heart and Aging Research in Genome Epidemiology) Consortium. Circulation 2010, 121, 1382–1392. [Google Scholar] [CrossRef]
  63. Heinrich, P.C.; Castell, J.V.; Andus, T. Interleukin-6 and the acute phase response. Biochem. J. 1990, 265, 621–636. [Google Scholar] [CrossRef]
  64. Yudkin, J.S.; Stehouwer, C.D.; Emeis, J.J.; Coppack, S.W. C-reactive protein in healthy subjects: Associations with obesity, insulin resistance, and endothelial dysfunction: A potential role for cytokines originating from adipose tissue? Arterioscler. Thromb. Vasc. Biol. 1999, 19, 972–978. [Google Scholar] [CrossRef]
  65. Zimmermann, E.; Anty, R.; Tordjman, J.; Verrijken, A.; Gual, P.; Tran, A.; Iannelli, A.; Gugenheim, J.; Bedossa, P.; Francque, S.; et al. C-reactive protein levels in relation to various features of non-alcoholic fatty liver disease among obese patients. J. Hepatol. 2011, 55, 660–665. [Google Scholar] [CrossRef]
  66. Del Bosque-Plata, L.; Martínez-Martínez, E.; Espinoza-Camacho, M.; Gragnoli, C. The Role of TCF7L2 in Type 2 Diabetes. Diabetes 2021, 70, 1220–1228. [Google Scholar] [CrossRef]
  67. Sladek, R.; Rocheleau, G.; Rung, J.; Dina, C.; Shen, L.; Serre, D.; Boutin, P.; Vincent, D.; Belisle, A.; Hadjadj, S.; et al. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 2007, 445, 881–885. [Google Scholar] [CrossRef] [PubMed]
  68. Hara, K.; Okada, T.; Tobe, K.; Yasuda, K.; Mori, Y.; Kadowaki, H.; Hagura, R.; Akanuma, Y.; Kimura, S.; Ito, C.; et al. The Pro12Ala polymorphism in PPAR gamma2 may confer resistance to type 2 diabetes. Biochem. Biophys. Res. Commun. 2000, 271, 212–216. [Google Scholar] [CrossRef]
  69. Lehmann, J.M.; Moore, L.B.; Smith-Oliver, T.A.; Wilkison, W.O.; Willson, T.M.; Kliewer, S.A. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma). J. Biol. Chem. 1995, 270, 12953–12956. [Google Scholar] [CrossRef]
  70. Knudsen, L.B.; Lau, J. The Discovery and Development of Liraglutide and Semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef]
  71. Osbak, K.K.; Colclough, K.; Saint-Martin, C.; Beer, N.L.; Bellanné-Chantelot, C.; Ellard, S.; Gloyn, A.L. Update on mutations in glucokinase (GCK), which cause maturity-onset diabetes of the young, permanent neonatal diabetes, and hyperinsulinemic hypoglycemia. Hum. Mutat. 2009, 30, 1512–1526. [Google Scholar]
  72. Dai, T.; Yang, Y.; Zhang, J.; Ma, X.; Chen, L.; Zhang, C.; Lv, S.; Li, L.; Tang, R.; Zhen, N.; et al. GCK exonic mutations induce abnormal biochemical activities and result in GCK-MODY. Front. Genet. 2023, 14, 1120153. [Google Scholar]
  73. Hattersley, A.T.; Turner, R.C.; Permutt, M.A.; Patel, P.; Tanizawa, Y.; Chiu, K.C.; O’Rahilly, S.; Watkins, P.J.; Wainscoat, J.S. Linkage of type 2 diabetes to the glucokinase gene. Lancet 1992, 339, 1307–1310. [Google Scholar] [CrossRef]
  74. Gidh-Jain, M.; Takeda, J.; Xu, L.Z.; Lange, A.J.; Vionnet, N.; Stoffel, M.; Froguel, P.; Velho, G.; Sun, F.; Cohen, D.; et al. Glucokinase mutations associated with non-insulin-dependent (type 2) diabetes mellitus have decreased enzymatic activity: Implications for structure/function relationships. Proc. Natl. Acad. Sci. USA 1993, 90, 1932–1936. [Google Scholar] [CrossRef]
  75. Lloyd, D.J.; St Jean, D.J., Jr.; Kurzeja, R.J.; Wahl, R.C.; Michelsen, K.; Cupples, R.; Chen, M.; Wu, J.; Sivits, G.; Helmering, J.; et al. Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors. Nature 2013, 504, 437–440. [Google Scholar] [CrossRef] [PubMed]
  76. Xie, Z.; Liu, J.; Xie, T.; Liu, P.; Hui, X.; Zhang, Q.; Xiao, X. Integration of proteomics and metabolomics reveals energy and metabolic alterations induced by glucokinase (GCK) partial inactivation in hepatocytes. Cell. Signal. 2024, 114, 111009. [Google Scholar]
  77. Ashton, K.S.; Andrews, K.L.; Bryan, M.C.; Chen, J.; Chen, K.; Chen, M.; Chmait, S.; Croghan, M.; Cupples, R.; Fotsch, C.; et al. Small molecule disruptors of the glucokinase-glucokinase regulatory protein interaction: 1. Discovery of a novel tool compound for in vivo proof-of-concept. J. Med. Chem. 2014, 57, 309–324. [Google Scholar] [CrossRef] [PubMed]
  78. De Ceuninck, F.; Kargar, C.; Ilic, C.; Caliez, A.; Rolin, J.O.; Umbdenstock, T.; Vinson, C.; Combettes, M.; de Fanti, B.; Harley, E.; et al. Small molecule glucokinase activators disturb lipid homeostasis and induce fatty liver in rodents: A warning for therapeutic applications in humans. Br. J. Pharmacol. 2013, 168, 339–353. [Google Scholar] [CrossRef]
  79. Borglykke, A.; Grarup, N.; Sparsø, T.; Linneberg, A.; Fenger, M.; Jeppesen, J.; Hansen, T.; Pedersen, O.; Jørgensen, T. Genetic variant SLC2A2 [corrected] Is associated with risk of cardiovascular disease—Assessing the individual and cumulative effect of 46 type 2 diabetes related genetic variants. PLoS ONE 2012, 7, e50418. [Google Scholar] [CrossRef] [PubMed]
  80. Teslovich, T.M.; Musunuru, K.; Smith, A.V.; Edmondson, A.C.; Stylianou, I.M.; Koseki, M.; Pirruccello, J.P.; Ripatti, S.; Chasman, D.I.; Willer, C.J.; et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 2010, 466, 707–713. [Google Scholar] [CrossRef]
  81. Varbo, A.; Benn, M.; Tybjærg-Hansen, A.; Grande, P.; Nordestgaard, B.G. TRIB1 and GCKR polymorphisms, lipid levels, and risk of ischemic heart disease in the general population. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 451–457. [Google Scholar] [CrossRef]
  82. Langer, S.; Jagdhuhn, D.; Waterstradt, R.; Gromoll, J.; Müller, M.; Rees, M.G.; Gloyn, A.L.; Baltrusch, S. Effects of coding variants in the glucokinase regulatory protein gene on hepatic glucose and triglyceride metabolism suggest a gene regulatory function of glucokinase. Metabolism 2025, 166, 156150. [Google Scholar]
  83. Zhang, Z.; Ji, G.; Li, M. Glucokinase regulatory protein: A balancing act between glucose and lipid metabolism in NAFLD. Front. Endocrinol. 2023, 14, 1247611. [Google Scholar]
  84. Cohen, J.C.; Boerwinkle, E.; Mosley, T.H., Jr.; Hobbs, H.H. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N. Engl. J. Med. 2006, 354, 1264–1272. [Google Scholar] [CrossRef] [PubMed]
  85. Doggrell, S.A.; Lynch, K.A. Is there enough evidence with evolocumab and alirocumab (antibodies to proprotein convertase substilisin-kexin type, PCSK9) on cardiovascular outcomes to use them widely? Evaluation of Sabatine MS, Giugliano RP, Wiviott SD et al. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events. N Engl J Med 2015;372:1500-1509, and Robinson JG, Farnier M, Krempf M et al. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med 2015;372:1488-99. Expert Opin. Biol. Ther. 2015, 15, 1671–1675. [Google Scholar] [CrossRef] [PubMed]
  86. Song, N.; Xu, H.; Liu, J.; Zhao, Q.; Chen, H.; Yan, Z.; Yang, R.; Luo, Z.; Liu, Q.; Ouyang, J.; et al. Design of a highly potent GLP-1R and GCGR dual-agonist for recovering hepatic fibrosis. Acta Pharm. Sin. B 2022, 12, 2443–2461. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, J.; Liu, S.; Wang, B.; Miao, Z.; Han, L.; Chu, N.; Zhang, K.; Meng, D.; Li, C.; Ma, X. Association between gout and polymorphisms in GCKR in male Han Chinese. Hum. Genet. 2012, 131, 1261–1265, Erratum in Hum. Genet. 2012, 131, 1267. [Google Scholar] [CrossRef]
  88. Matsuo, H.; Yamamoto, K.; Nakaoka, H.; Nakayama, A.; Sakiyama, M.; Chiba, T.; Takahashi, A.; Nakamura, T.; Nakashima, H.; Takada, Y.; et al. Genome-wide association study of clinically defined gout identifies multiple risk loci and its association with clinical subtypes. Ann. Rheum. Dis. 2016, 75, 652–659. [Google Scholar] [CrossRef]
Figure 1. The biological functions related to GKRP. Glucokinase regulatory protein (GKRP) is located at the nucleus of the hepatocyte. When the serum glucose increases, GKRP releases GCK and enables the migration of GCK to the cytoplasm, where it converts glucose to glucose-6-P, which can be stored as glycogen or as fat (through de novo lipogenesis). Meanwhile, the enhancement of glycolysis leads to increased purine metabolism and releases more urate into the blood, causing gout. On the other hand, fructose 1-phosphate (fructose-1-P) is a natural disruptor of the GKRP-GCK complex, whereas fructose 6-phosphate (fructose-6-P) stimulates its binding. Abbreviations: GKRP, glucokinase regulatory protein; GCK, glucokinase; ATP, adenosine triphosphate; ADP, adenosine diphosphate; AMP, adenosine monophosphate; IMP, inosine monophosphate.
Figure 1. The biological functions related to GKRP. Glucokinase regulatory protein (GKRP) is located at the nucleus of the hepatocyte. When the serum glucose increases, GKRP releases GCK and enables the migration of GCK to the cytoplasm, where it converts glucose to glucose-6-P, which can be stored as glycogen or as fat (through de novo lipogenesis). Meanwhile, the enhancement of glycolysis leads to increased purine metabolism and releases more urate into the blood, causing gout. On the other hand, fructose 1-phosphate (fructose-1-P) is a natural disruptor of the GKRP-GCK complex, whereas fructose 6-phosphate (fructose-6-P) stimulates its binding. Abbreviations: GKRP, glucokinase regulatory protein; GCK, glucokinase; ATP, adenosine triphosphate; ADP, adenosine diphosphate; AMP, adenosine monophosphate; IMP, inosine monophosphate.
Cimb 47 00850 g001
Figure 2. The pathogenic mechanism of P446L-GKRP. The GCKR variant rs1260326 leads to GKRP P446L and promotes de novo lipogenesis, leading to hepatic fat deposition and hypertriglyceridemia. P446L-GKRP disturbs the production of the GKRP-GCK complex and enables GCK to migrate into the cytoplasm. The increased glycolysis produces more pyruvate, which results in increased acetylCoA production. AcetylCoA participates in the citric acid cycle, leading to higher amounts of citrate. Excessive citrate is then used to synthesize fatty acids. MalonyCoA produced in the process of de novo lipogenesis hampers the oxidation of fatty acids (FAs) by inhibiting CPT1, a mitochondrial FA transporter, which leads to liver fat deposition. Meanwhile, increased fatty acid formation results in TG accumulation and increased hepatic fat, stimulating the production of ApoB VLDL particles, which are degraded into IDL and LDL and release more TGs. HDL and LDL particles exchange cholesteryl ester (CE) for TGs with VLDL particles, mediated by CETP. The above may result in higher serum triglyceride levels and lead to hypertriglyceridemia. Abbreviations: GKRP, glucokinase regulatory protein; GCK, glucokinase; CoA, coenzyme A; CPT1, Carnitine palmitoyltransferase1; TG, triglyceride; ApoB, apolipoproteinB; VLDL, very-low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; CETP, cholesteryl ester transfer protein; HDL, high-density lipoprotein.
Figure 2. The pathogenic mechanism of P446L-GKRP. The GCKR variant rs1260326 leads to GKRP P446L and promotes de novo lipogenesis, leading to hepatic fat deposition and hypertriglyceridemia. P446L-GKRP disturbs the production of the GKRP-GCK complex and enables GCK to migrate into the cytoplasm. The increased glycolysis produces more pyruvate, which results in increased acetylCoA production. AcetylCoA participates in the citric acid cycle, leading to higher amounts of citrate. Excessive citrate is then used to synthesize fatty acids. MalonyCoA produced in the process of de novo lipogenesis hampers the oxidation of fatty acids (FAs) by inhibiting CPT1, a mitochondrial FA transporter, which leads to liver fat deposition. Meanwhile, increased fatty acid formation results in TG accumulation and increased hepatic fat, stimulating the production of ApoB VLDL particles, which are degraded into IDL and LDL and release more TGs. HDL and LDL particles exchange cholesteryl ester (CE) for TGs with VLDL particles, mediated by CETP. The above may result in higher serum triglyceride levels and lead to hypertriglyceridemia. Abbreviations: GKRP, glucokinase regulatory protein; GCK, glucokinase; CoA, coenzyme A; CPT1, Carnitine palmitoyltransferase1; TG, triglyceride; ApoB, apolipoproteinB; VLDL, very-low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; CETP, cholesteryl ester transfer protein; HDL, high-density lipoprotein.
Cimb 47 00850 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, K.; Chen, P.; Su, Y.; Chen, Y.; Song, X.; Yu, B.; Wang, H. Significant Association Between Glucokinase Regulatory Protein Variants and Genetic and Metabolic Diseases. Curr. Issues Mol. Biol. 2025, 47, 850. https://doi.org/10.3390/cimb47100850

AMA Style

Xu K, Chen P, Su Y, Chen Y, Song X, Yu B, Wang H. Significant Association Between Glucokinase Regulatory Protein Variants and Genetic and Metabolic Diseases. Current Issues in Molecular Biology. 2025; 47(10):850. https://doi.org/10.3390/cimb47100850

Chicago/Turabian Style

Xu, Ke, Peng Chen, Yujing Su, Yanghui Chen, Xiuli Song, Bo Yu, and Hong Wang. 2025. "Significant Association Between Glucokinase Regulatory Protein Variants and Genetic and Metabolic Diseases" Current Issues in Molecular Biology 47, no. 10: 850. https://doi.org/10.3390/cimb47100850

APA Style

Xu, K., Chen, P., Su, Y., Chen, Y., Song, X., Yu, B., & Wang, H. (2025). Significant Association Between Glucokinase Regulatory Protein Variants and Genetic and Metabolic Diseases. Current Issues in Molecular Biology, 47(10), 850. https://doi.org/10.3390/cimb47100850

Article Metrics

Back to TopTop