Review Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Feb 15, 2024; 15(2): 142-153
Published online Feb 15, 2024. doi: 10.4239/wjd.v15.i2.142
Genotype-based precision nutrition strategies for the prediction and clinical management of type 2 diabetes mellitus
Omar Ramos-Lopez, Medicine and Psychology School, Autonomous University of Baja California, Tijuana 22390, Baja California, Mexico
ORCID number: Omar Ramos-Lopez (0000-0002-2505-1555).
Author contributions: Ramos-Lopez O contributed to the writing and revision of this manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
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: Omar Ramos-Lopez, PhD, Professor, Medicine and Psychology School, Autonomous University of Baja California, Universidad 14418, UABC, Parque Internacional Industrial Tijuana, Tijuana 22390, Baja California, Mexico. oscar.omar.ramos.lopez@uabc.edu.mx
Received: October 14, 2023
Peer-review started: October 14, 2023
First decision: December 6, 2023
Revised: December 7, 2023
Accepted: January 11, 2024
Article in press: January 11, 2024
Published online: February 15, 2024

Abstract

Globally, type 2 diabetes mellitus (T2DM) is one of the most common metabolic disorders. T2DM physiopathology is influenced by complex interrelationships between genetic, metabolic and lifestyle factors (including diet), which differ between populations and geographic regions. In fact, excessive consumptions of high fat/high sugar foods generally increase the risk of developing T2DM, whereas habitual intakes of plant-based healthy diets usually exert a protective effect. Moreover, genomic studies have allowed the characterization of sequence DNA variants across the human genome, some of which may affect gene expression and protein functions relevant for glucose homeostasis. This comprehensive literature review covers the impact of gene-diet interactions on T2DM susceptibility and disease progression, some of which have demonstrated a value as biomarkers of personal responses to certain nutritional interventions. Also, novel genotype-based dietary strategies have been developed for improving T2DM control in comparison to general lifestyle recommendations. Furthermore, progresses in other omics areas (epigenomics, metagenomics, proteomics, and metabolomics) are improving current understanding of genetic insights in T2DM clinical outcomes. Although more investigation is still needed, the analysis of the genetic make-up may help to decipher new paradigms in the pathophysiology of T2DM as well as offer further opportunities to personalize the screening, prevention, diagnosis, management, and prognosis of T2DM through precision nutrition.

Key Words: Type 2 diabetes mellitus, Nutrigenetics, Single nucleotide polymorphism, Genotype, Diet, Precision nutrition

Core Tip: The onset and progression of type 2 diabetes mellitus (T2DM) is influenced by complex interrelationships between genetic and dietary factors. Indeed, a number of nutrigenetic studies have identified significant gene-diet interactions related to T2DM predisposition, nutrient metabolic status, and dietary intervention responsiveness. Moreover, this knowledge has motivated the interest for the design and implementation of genotype-based dietary strategies for improving glycemic outcomes compared to conventional nutritional advice. Although more investigation is required, these insights may help to explain disease phenotype heterogeneity, with relevance in precision nutrition for the personalized prevention and clinical management of T2DM.



INTRODUCTION

Type 2 diabetes mellitus (T2DM) is a metabolic disease caused by insufficient pancreatic insulin secretion or defective hormone actions in target tissues[1]. T2DM is recognized as a major public health concern due to rising global prevalence and negative impact on human wellbeing and life expectancy, being significantly associated with morbidity burden and premature mortality[2].

Several factors have been identified to contribute to the prevalence of T2DM including the genetic background[3]. Accordingly, a number of sequence DNA variants across the human genome have been characterized, some of which may affect gene expression and protein functions relevant for maintaining glucose homeostasis[3-5]. Largely, single nucleotide polymorphisms (SNPs) have been the most prevalent studied genetic variations in the field of precision medicine, with applications in T2DM prevention and personalized management[6-8]. Moreover, genetic risk scores (GRS) have been developed to assess the additive effect of SNPs[9-11].

Of note, the genetic contribution to T2DM status may depend on interactions with environmental issues including diet, which may explain some of the inconsistencies reported among epidemiological studies relating diet to chronic diseases[12]. Thus, interrelationships between genetic variants and dietary features (i.e., intakes of macro and micronutrients, eating behaviors, nutritional patterns, and the consumption of particular foods) may influence T2DM risk or disease complications by affecting critical pathways involved in glucose signaling, insulin secretion, β-cell function, gluco-lipotoxicity, inflammation and oxidative stress[12-14]. Therefore, people with higher genetic predisposition should avoid certain harmful foods or adopt healthy dietary patterns to delay T2DM onset.

In this context, it has been illustrated that the combination of genetic (52 SNPs in 37 genes) and dietary data (food with high sugar content) using machine learning approaches may improve the prediction of T2DM incidence[15]. Likewise, high genetic (48 SNPs) and dietary risk scores (based on sugar-sweetened beverages, processed meat, whole grains and coffee) were associated with increased incidence of T2DM[16].

In this document, potential interactions between genetic polymorphisms and dietary factors concerning T2DM susceptibility and disease progression are reviewed, some of which have demonstrated a value as biomarkers of personal responses to nutritional interventions. Also, novel genotype-based dietary strategies for the prevention and clinical management of T2DM are documented. Future directions comprising the integration of genetics with another omics tools are also postulated. These insights may help to explain heterogeneity in predisposition to T2DM and the development of related systemic complications, with relevance in disease stratification and precision nutrition through the study of the human genome.

GENETIC BACKGROUND, DIETARY INTAKE, AND T2DM RISK

A relevant precision nutrition approach in T2DM risk prediction/prevention include the analysis of associations between genetic polymorphisms and T2DM that are modulated by dietary features. Indeed, a number of nutrigenetic studies have identified significant gene-diet interactions related to T2DM predisposition (Table 1). These include single SNPs mapped to genes involved in pivotal physiological processes such as energy breakdown, nutrient utilization, insulin signaling, circadian rhythm, cell cycle regulation, pancreatic function, hypothalamic food intake control, neuronal synapse, signal transduction, and taste perception, which interact with nutritional factors to influence T2DM risk (Table 1). Among them, the consumption of particular foods (vegetables, whole grains, coffee, olive oils, alcoholic beverages, and dairy products), macronutrients (carbohydrates, fatty acids, protein, fiber) and micronutrients (iron, folate) intakes, adherence to dietary patterns, and eating time schedules (Table 1).

Table 1 Gene-diet interactions concerning the risk of developing type 2 diabetes mellitus and individual responses to nutritional interventions.
SNP reference
Gene symbol
Gene function
Risk allele
Dietary interaction
Main outcome
Population
Ref.
rs7903146TCF7L2Wnt signaling pathwayTHigh dessert and milk intakes (above median)Higher T2DM riskAlgerian[83]
rs7903146TCF7L2Wnt signaling pathwayCFiber intakeInversely associated with T2DM incidenceSwedish[84]
rs7903146 and rs4506565TCF7L2Wnt signaling pathwayrs7903146 (C) and rs4506565 (A)Per daily 30-g increased intake of whole grain and per daily 5-g increased intake of cereal fiberDecreased risk of developing T2DMSwedish men[85]
rs7901695TCF7L2Wnt signaling pathwayTUpper protein intake quantilesHigher HbA1c, HOMA-IR, blood glucose, and insulin levelsPolish[86]
rs6696797, rs4244372, and rs10881197AMY1Carbohydrate digestionrs6696797 (A), rs4244372 (A), rs10881197 (G)Carbohydrate intake > 65% of total energyHigher T2DM incidenceKorean women[87]
rs2233998TAS2R4Bitter taste perceptionTHigh intakes of carbohydrates or sugars (highest tertile) and low intakes of fruits or vegetables (lowest tertile)Higher T2DM incidenceKorean women[88]
rs1801282 and rs3856806PPARGFatty acid storage and glucose metabolismrs1801282 (Pro12), rs3856806 (C)High fat consumption (the third sex-specific tertile of fat intakeIncreased T2DM riskFrench[89]
rs7756992CDKAL1Beta cells functionGFirst tertiles of protein and fat intakesHigher T2DM riskKorean[90]
rs7754840CDKAL1Pancreatic beta cells functionGHabitual coffee intakeLower risk of prediabetes and T2DMEast Asians[91]
rs5215KCNJ11Formation of ATP-sensitive potassium (K-ATP) channels in pancreatic beta cellsCHabitual coffee intakeLower risk of prediabetes and T2DMEast Asians[91]
rs4402960IGF2BP2Cellular metabolism modulation by post transcriptional regulationTHabitual coffee intakeLower risk of prediabetes and T2DMEast Asians[91]
rs10517030PGC-1αRegulation of genes involved in energy metabolismCLow-energy diet (daily consumption less than estimated energy intake)Positively associated with T2DM prevalence and insulin resistance and negatively associated with beta cell functionKoreans[92]
rs6265BDNFSurvival and growth of neurons, and synaptic efficiency and plasticityMetLow-energy (daily consumption less than estimated daily energy intake), low-protein (< 13% daily energy), and high-carbohydrate (70% daily energy)Lower risk for T2DMKoreans[93]
rs161364 and rs8065080TRPV1Receptor for capsaicin, non-selective cation channel, and participates in transduction of painful thermal stimulirs161364 (T) and rs8065080 (C)High preference for oily foods and high fat intake from oily foodsLower risk for T2DMKoreans[94]
rs77768175, rs2074356 and rs11066280HECTD4Glucose homeostasis and glucose metabolic processrs77768175 (A), rs2074356 (G), rs11066280 (T)Alcohol consumption (> 5 g/d)Significantly increased risks of T2DMEast Asians[95]
rs10830963MTNR1BRegulation of the circadian actions of melatoninGIncreasing dietary iron intakeIncreased risk of elevated fasting glucose, higher fasting glucose, and higher HbA1cChinese[96]
rs10830963MTNR1BRegulation of the circadian actions of melatoninGLate dinnerImpaired glucose toleranceEuropean[97]
rs10830963MTNR1BRegulation of the circadian actions of melatoninGLate eatingImpaired glucose tolerance and insulin secretion defectsEuropean[98]
rs2943641IRS1Insulin signalingTLower tertiles of carbohydrate intake (women) and lowest tertile of fat intake (men)Decreased risk of T2DMSwedish[99]
rs7578326 and rs2943641IRS1Insulin signalingrs7578326 (G) and rs2943641 (T)Low SFA-to-carbohydrate ratio (≤ 0.24)Lower risk of insulin resistance and metabolic syndromeAmerican[100]
rs10423928GIPRInsulin release stimulationTHighest carbohydrate quintileDecreased T2DM riskSwedish[101]
rs3014866S100A9Cell cycle progression and differentiationCHigh dietary SFA: Carbohydrate ratio intakeHigher insulin resistanceSpanish white adults, North American non-Hispanic white adults, and Hispanic adults[102]
rs709592PSMD3Maintenance of protein homeostasisTLow carbohydrate intake (≤ 49.1% energy)Higher insulin resistanceAmericans[103]
rs8065443PSMD3Maintenance of protein homeostasisALow (n-3):(n-6) PUFA ratio (≤ 0.11)Higher insulin resistanceAmericans[103]
rs7645550KCNMB3Control of smooth muscle tone and neuronal excitabilityTLow (n-3):(n-6) PUFA ratio (≤ 0.11)Lower insulin resistanceAmericans[104]
rs1183319KCNMB3Control of smooth muscle tone and neuronal excitabilityGHigh (n-3):(n-6) PUFA ratio (> 0.09)Higher HbA1c levelsHispanics[104]
rs2270188CAV2Signal transduction, lipid metabolism, cellular growth control and apoptosisTIncrease of daily fat intake from 30% to 40% energyGreater risk of T2DMEuropean[105]
rs10923931NOTCH2Wnt signaling pathwayTIncreasing fiber intakeLower T2DM riskSwedish[106]
rs4457053ZBED3Wnt signaling pathwayGIncreasing fiber intakeLower T2DM riskSwedish[106]
rs3765467GLP1RInsulinotropic action of GLP-1 in β-cellsGHighest tertiles of energy, protein and carbohydrate consumptionHigher risk for decreased insulin secretionJapanese men[107]
rs9939609FTORegulation of energy intakeALow adherence to the Mediterranean diet (≤ 9 points)Higher risk of prevalent T2DMSpanish[108]
rs9939609FTORegulation of energy intakeALow folate intake (< 406 μg/d)Higher fasting plasma glucose concentrationsSpanish[108]
rs17782313MC4RHypothalamic leptin-melanocortin signaling pathwayCLow adherence to the Mediterranean diet (≤ 9 points)Higher risk of prevalent T2DMSpanish[108]

In addition, GRS have been constructed to evaluate the cumulative effects of SNPs on T2DM susceptibility, where dietary factors are implicated. For instance, and obesity GRS positively interacted with dietary intake of cholesterol to affect insulin resistance in overweight/obese Spanish individuals[17]. Of note, Brazilian subjects with high GRS for metabolic disease and total fat intakes had increased blood glucose and insulin-related traits than those with low GRS[18]. Conversely, lower serum levels of glycated hemoglobin were found in Ghanaian adults with low total fat intake (≤ 36.5 g/d) despite carrying more than two risk alleles of vitamin D-related genetic variants[19]. Also, associations between a GRS related to insufficient glucose-stimulated insulin secretion and T2DM risk was accentuated in Asian individuals with high energy and calcium intakes[20]. Moreover, Korean subjects carrying polygenic variants linked to oxidative stress had increased risk of T2DM, which was lowered the by the intakes of dietary antioxidants[21]. Besides, the genetic predisposition to T2DM was exacerbated with higher intakes of dietary branched-chain amino acids in Chinese[22].

Regarding specific foods, it was reported that middle-aged Korean adults with high GRS affecting insulin signaling presented more instances of insulin resistance when combined with high coffee (≥ 10 cups/wk) or caffeine (≥ 220 mg/d) intakes[23]. Likewise, alcohol consumption significantly increased the risk of T2DM especially in Chinese men with low genetic predisposition to insulin secretion deterioration[24]. In the same way, the association between the consumption of sugar-sweetened beverages and serum glucose abnormalities was stronger in Chileans with high T2DM genetic susceptibility[25]. Conversely, augmented genetic risk for T2DM was ameliorated by increasing the consumption of fruits in Chinese population[26]. In line with this finding, lower plant protein intake (< 39 g/d) was identified as a factor contributing to increase the risk of T2DM in genetically predisposed Asian Indians[27].

Furthermore, a high GRS for impaired insulin secretion increased the risk of T2DM by consuming a low-carbohydrate Western dietary pattern in Korean adults[28]. In Asians, higher fasting serum glucose concentrations were found in participants with high T2DM-linked GRS who adopted a Western dietary pattern[29]. On the contrary, it was reported that Koreans with high GRS for insulin resistance may be benefited by consuming a plant-based diet with high amounts of fruits, vitamin C, and flavonoids[30].

These studies show evidence concerning interactions between genetic variants and T2DM risk depending on dietary intakes, which may be useful for the design of nutritional therapies aimed to control the burden of T2DM, although more research is needed in populations with different genetic ancestries including Hispanics and Africans.

GENE-DIET INTERACTIONS AFFECTING METABOLIC STATUS IN T2DM PATIENTS

Once T2DM has established, several physiopathological processes affecting glucose/lipid metabolism homeostasis, immune function, adipokine secretion, and gut microbiota dysbiosis play a critical role in the development of vascular injuries including diabetic heart disease and stroke[31]. Thus, it is important to monitor the metabolic status in T2DM in order to prevent or delay the progression of complications associated with this disease.

Accordingly, some studies have analyzed the effect of gene-diet interactions on glycemic, lipid, and inflammatory features in T2DM patients, with relevance in clinical disease management. In this regard, studies in Mexican population have evidenced relevant gene-nutrient interactions concerning glycemic control and lipid profile in T2DM. For example, positive correlations were found between calcium intake and glycated hemoglobin and potassium intake and triglyceride-glucose index only in carriers of the 408 Val risk allele of the SLC22A1/OCT1 Met408Val polymorphism[32]. Also, higher blood concentrations of total cholesterol, non-high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol were found in carriers of the APOE ε2 allele with low consumption of monounsaturated fatty acids (MUFA), whereas carriers of the apolipoprotein E (APOE) ε4 allele with high dietary ω-6:ω-3 polyunsaturated fatty acids (PUFA) ratio presented higher glycated hemoglobin levels[33]. Likewise, A1 allele carriers of the DRD2/ANKK1 TaqIA polymorphism were protected from serum triglyceride increases by maltose intake, but A2A2 homozygotes were susceptible to triglyceride rises through excessive consumptions of total fat, MUFA, and dietary cholesterol[34].

In Iranians with T2DM, Met allele carriers of the brain-derived neurotrophic factor (BDNF) Val66Mat polymorphism with high scores of dietary indices showed lower blood levels of triglycerides ((healthy eating index and diet quality index), total cholesterol, and interleukin-18 (phytochemical index) than Val/Val homozygotes[35]. Meanwhile, C-allele carriers of the APOA2-265 T>C polymorphism had highest means of body mass index, waist circumference, blood cholesterol and serum ghrelin and leptin levels when dietary acid load (either potential renal acid load or net endogenous acid production) values were high[36]. Of note, higher inflammatory and antioxidant markers including C-reactive protein, total antioxidant capacity, superoxide dismutase, and 8-isoprostaneF2alpha were found in B2B2 homozygotes of the CETP TaqB1 polymorphism when they consumed diets with high dietary insulin index[37]. Similarly, risk-allele carriers (CG, GG) of the peroxisome proliferator-activated receptor (PPAR) Pro12Ala polymorphism who consumed a diet with high dietary insulin load and insulin indexes were more likely to be obese and have increased inflammatory markers (i.e., interleukin-18, isoprostaneF2α, and pentraxin-3) compared to individuals with the CC genotype[38]. Moreover, worse plasma lipid profile was found in participants carrying the AA/AG genotype of the ApoB EcoRI polymorphism when increasing the percentage of energy derived from dietary fat, carbohydrates, protein, saturated fatty acids (SFA), and cholesterol in comparison to GG homozygotes[39]. In the same way, Del-allele carries of the ApoB Ins/Del genetic variant who consumed high amounts of MUFA (≥ 12% E) and carbohydrates (≥ 54% E) had higher blood levels of triglycerides and low density lipoprotein-cholesterol, while low carbohydrate (< 54% E) intakes were associated with raised serum concentrations of leptin and ghrelin in T2DM patients with this same genetic profile compared to Ins/Ins homozygotes[40]. In addition, an increased risk of obesity was found in carriers of the Del allele of ApoB gene when combined with a low consumption of dietary ω-3 PUFA (< 0.6% E) in T2DM subjects[41]. Taken together, these results could be useful to prevent cardiometabolic risk factors and later complications in T2DM patients via manipulation of dietary intakes of selected nutrients mainly in genetically susceptible individuals. However, more investigation is needed in other populations with diverse ancestries and exposed to different environments in order to regionalize antidiabetic nutritional treatments.

GENETIC POLYMORPHISMS AS BIOMARKERS OF GLYCEMIC RESPONSES TO DIETARY ADVICE

Dietary strategies aimed to achieve or improve glucose homeostasis not always have a positive impact in all individuals, which can be due to genetic factors. In this sense, some trials have evaluated the value of SNPs as potential biomarkers of glycemic outcomes in response to different nutritional interventions. For instance, the variant rs3071 of the SCD gene modified blood glucose response to dietary oils varying in MUFA content in adults with obesity, where CC genotype carriers showed an increase in blood glucose levels with a high SFA/low MUFA control oil, but reductions in this outcome with both high MUFA oil diets[42]. Within the multicenter NUGENOB study, the T allele of the protein phosphatase Mg(2+)/Mn(2+)-dependent 1K (PPM1K) rs1440581 genetic variant was associated with higher reductions of serum insulin and homeostasis model assessment (HOMA)-B after a high-fat (40%-45% E) diet, whereas an opposite effect was found in the low-fat (20%-25% E) diet group[43]. Also, obese individuals who were homozygous for the T-risk allele of the transcription factor 7 like 2 (TCF7L2) rs7903146 polymorphism and consumed a high-fat (40%-45% E) diet, underwent smaller reductions in HOMA-estimated insulin resistance (HOMA-IR)[44].

Findings from the POUNDS lost trial revealed greater decreases in fasting glucose, serum insulin, and HOMA-IR in T-allele participants of the glucose-dependent insulinotropic polypeptide receptor (GIPR) rs2287019 variant who were assigned to low-fat (20%-25% E) diets[45]. In addition, subjects with the risk-conferring CC genotype of the insulin receptor substrate-1 (IRS1) rs2943641 SNP had greater decreases in insulin and HOMA-IR than those without this genetic profile in the highest-carbohydrate (65% E) dietary group[46]. Whereas, the T allele of deficient activity of 7-dehydrocholesterol reductase (DHCR7) rs12785878 polymorphism was associated with higher decreases in serum insulin and HOMA-IR only in high-protein (25% E) diets[47]. Similarly, greater drops in fasting insulin levels were related to the PCSK7 rs236918 G allele in high-dietary carbohydrate (65% E) intakes, especially in white Americans[48]. Of note, carriers of the risk allele (A) of the Fat mass and obesity associated (FTO) rs1558902 variant benefited more in improving insulin sensitivity by consuming high-fat (40%-45% E) diets rather than low-fat (20%-25% E) regimens[49].

In a Spanish cohort with obesity, improvements in serum insulin levels and HOMA-IR were associated with the ADRB3 Trp64Trp genotype after hypocaloric diet with high protein (34% E) content[50]. Besides, AA genotype carries of the BDNF rs10767664 variant underwent reductions in insulin resistance markers when consumption of MUFA (67.5%) was high[51]. Likewise, TNFA-308GG homozygotes had a better glycemic response after high (22.7%) dietary intakes of PUFA[52]. In the same say, UCP3 55CC genotype carriers benefited more (more decreases in blood glucose, serum insulin, and HOMA-IR) when consumed a high-protein (34% E) diet[53]. Interestingly, it was suggested that the T allele of the ADIPOQ rs1501299 SNP was related to a lack of response of fasting glucose/insulin and HOMA-IR secondary to a Mediterranean-style diet in Spanish obese individuals[54]. Insulin resistance was ameliorated after the consumption of this same dietary pattern in T allele carries of the RETN rs10401670 gene polymorphism[55]. Comparable results were reported concerning insulin resistance reductions in CC genotype carries of the melatonin receptor 1B (MTNR1B) rs10830963 variant but not in GC + GG groups after following a hypocaloric diet with Mediterranean pattern[56].

Some studies have evaluated the cumulative effect of multiple SNPs (by calculating GRS) instead of single variants. In this context, participants with high genetic risk of glucose abnormalities showed increased fasting glucose after consuming a high-fat diet (40%-45% E), which was not observed in subjects assigned to the low-fat (20%-25% E) group[57]. A lower GRS for diabetes was associated with higher reductions in fasting insulin, glycated hemoglobin, and HOMA-IR, and a lesser increase in HOMA-B only when the consumption of dietary protein (15% E) was low[58]. In the meantime, insulin resistance improvements were limited to individuals with a higher GRS of habitual coffee consumption following a low-fat (20%-25% E) dietary intervention[59].

The influence of the genetic background on metabolic outcomes after dietary treatments have also been assessed in T2DM patients. For example, a dietary intervention based on increased intakes of whole grains, vegetables, and legumes was able to prevent an age-related increase in blood triglyceride concentrations in Koreans with impaired fasting glucose or new-onset of T2DM carrying the TT genotype of the APOA5-1131 T>C SNP[60]. Accordingly, low glycemic index diets induced significant decreases of serum lipids, fasting blood glucose, and glycated albumin only in Chinese women with T2DM who were FABP2 Ala54 homozygotes[61]. Furthermore, carriers of the FTO rs9939609 risk allele (A) underwent a better response in improving body mass index and diastolic blood pressure in response to supplementation with epigallocatechin-3-gallate (300 mg/d) in Iranian patients with T2DM[62].

Overall, current evidence suggests a role of selected genetic polymorphisms in modulating the individual metabolic responses to some dietary treatments. However, available studies have been performed mainly in Europeans/Caucasians, with particular genetic backgrounds; therefore, additional studies in different populations are required including Latin Americans, Africans, and Asians. Also, the analysis of the effects of supplementation with antioxidant micronutrients and bioactive compounds with anti-inflammatory properties is warranted.

GENOTYPE-BASED DIETARY INTERVENTIONS AND GLYCEMIC OUTCOMES

The knowledge about the implication of genetic variants and dietary factors in the onset and progression of T2DM has motivated the interest for the design and implementation of genotype-based intervention strategies for improving glycemic/metabolic outcomes compared to traditional nutritional prescriptions. For instance, it was evidenced that a personalized low-glycemic index nutrigenetic diet (utilizing 28 SNPs with evidence of gene-diet/lifestyle interactions) induced higher fasting glucose reductions than a Ketogenic diet in overweight/obese individuals[63]. Likewise, healthier effects in HOMA-IR and insulin serum levels were observed in MTHFR 677T allele carriers consuming a GENOMEX diet comprising of diet-related adaptive gene polymorphisms highly prevalent in Mexicans[64]. However, no differences were detected regarding glucose homeostasis outcomes at 24 wk of follow-up between a nutrigenetic-guided diet (using genetic information of a proprietary algorithm) and a standard balanced diet in obese or overweight American veterans[65].

In T2DM patients, a case study based on the N-of-1 approach revealed better glycemic control when adhered to a genetically-guided Mediterranean diet (high-quality foods rich in fiber and antioxidants that have been proven to exert beneficial glycaemia effects) considering genetic variants guiding the personalized selection of macronutrients for the nutritional management of T2DM[66]. Similarly, greater improvements in fasting plasma glucose and glycosylated hemoglobin concentrations were found in patients with pre-diabetes or T2DM following a personalized nutritional plan (taking in consideration SNPs associated with individual responses to macronutrient intakes) compared to conventional medical nutrition therapy[67].

Furthermore, some studies have evaluated the utility of genetic disclosure as a tool for T2DM prevention and disease control. For example, participants who received diabetes genetic risk counseling together with general education about modifiable risk factors and personal stimulus to adopt diabetes lifestyle prevention behaviors reported high levels of support, perceived personal control and satisfaction with the genetic counseling sessions[68]. Nevertheless, diabetes genetic risk testing and counseling did not necessarily improved disease prevention behaviors such as self-reported motivation or prevention program adherence among overweight individuals at increased phenotypic risk for T2DM[69]. Moreover, comparison analyzes did not revealed significant differences between genetic testing results and traditional risk counseling concerning behavior changes to reduce the risk of T2DM in non-diabetic overweight/obese veterans[70]. Given inconsistences in available evidence, more research is needed to translate this knowledge into clinical care in T2DM. Further investigation should contemplate information that could interfere with the results including the prevalence and metabolic effects of selected SNPs, cultural level of populations, compatibility of dietary plans with genotypic characteristics, and the quality of nutritional/lifestyle advice.

FUTURE DIRECTIONS

In addition to genetics, progresses in other omics areas are improving current understanding of the biological/molecular mechanisms involved in T2DM pathogenesis and clinical outcomes[71]. Similar to the influence of the genetic background, it has been evidenced that epigenetic modifications may alter transcriptional activity resulting in different T2DM traits and phenotypes; certainly, different genes responsible for the interindividual variability in responses to antidiabetic treatments (including dietary advice) are subjected to epigenetic regulation[72]. More importantly, interactions among polymorphisms in key metabolic genes (i.e., TCF7L2), related methylation status, and environmental factors have been suggested as a possible etiologic pattern for T2DM[73]. Besides, SNPs in microRNA (miRNA) genes may change the structure of miRNAs and their target gene expressions to influence T2DM risk[74].

Also, metagenomic and metabolomic methodologies have emerged to investigate the interrelationships between the gut microbiota dysbiosis and their related metabolites (affecting critical metabolic pathways in the host such as immunity and nutrient metabolism) in the development of T2DM[75]. Of note, characterization of gut microbiota of individuals carrying the risk alleles of the PPARGC1A (rs8192678) and PPARD (rs2267668) variants revealed some taxa (with overrepresentation of ABC sugar transporters) putatively associated with insulin resistance and T2DM[76]. Correspondingly, the MMP27 rs7129790 polymorphism was strongly associated with high gut abundance of Proteobacteria in Mexican Americans with a high prevalence of obesity and T2DM[77].

Moreover, high-throughput proteomics assays have allowed the discovery and representation of potential protein-T2DM links, providing novel intervention targets in this disease[78]. Interestingly, a set of circulating proteins causally associated with T2DM were identified using two-sample Mendelian randomization approaches, which is a validated method to examine the causal effect of variation in genes of known function on disease[79]. Also, Mendelian randomization analyses did not uncover significant causal effects between proteins (i.e., retinal dehydrogenase 1, galectin-4, cathepsin D, and lipoprotein lipase) and diabetes, suggesting that identified proteins are expected to be biomarkers for T2DM, rather than demonstrating causal pathways[80].

Additionally, coupling genomic data (i.e., GRS) with conventional phenotypical information (i.e., age, sex, body composition, medication use, and vital signs) is being useful for enhancing individual T2DM risk stratification and disease prediction[81,82]. Advances in next-generation sequencing technologies and the use of machine learning and other artificial intelligence methods became fundamental to analyze these T2DM-associated multiomics datasets.

CONCLUSION

Current evidence support the impact of genetic variation on the risk of developing blood glucose/insulin alterations and subsequent T2DM as well as its implication in affecting the lipid, inflammatory, and carbohydrate status in T2DM patients through interactions with dietary factors. These include SNPs and other structural variants mapped to metabolically active genes such as TCF7L2, amylase 1, TAS2R4, PPARG, CDKAL1, KCNJ11, insulin-like growth factor 2 binding protein 2, proliferator-activated receptor-gamma coactivator-1alpha, BDNF, transient receptor potential vanilloid-1 channel, HECT domain E3 ubiquitin protein ligase 4, MTNR1B, IRS1, GIPR, S100A9, PSMD3, KCNMB3, Caveolin-2, NOTCH2, zinc finger BED-type containing 3, GLP1R, FTO, melanocortin 4 receptor, SLC22A1/OCT1, APOE, DRD2/ANKK1, APOA2, CETP, PPAR-γ, and ApoB, which have been analyzed using single and cumulative approaches. Moreover, some genetic polymorphisms have been identified as putative biomarkers of individual responses to energy-restricted nutritional prescriptions aimed to glucose control including those located in SCD, PPM1K, FTO, TCF7L2, GIPR, IRS1, DHCR7, PCSK7, ADRB3, BDNF, TNFA, UCP3, ADIPOQ, RETN, MTNR1B, APOA5, and FABP2 genes. Furthermore, some genotype-based dietary strategies have been developed for improving T2DM control in comparison to general lifestyle recommendations for all people. However, more research is needed in order to expand and confirm these findings in other populations less explored such as Latin Americans and Africans considering some sources of variability (i.e., allele frequency, quantitative trait locus, and gender influence) incorporating the assessment of the role of food bioactive compounds and micronutrients in prospective dietary interventions. In any case, the analysis of the genetic make-up may help to decipher new paradigms in the pathophysiology of T2DM as well as offer further opportunities to personalize the screening, prevention, diagnosis, management, and prognosis of T2DM.

Footnotes

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

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: Mexico

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Horowitz M, Australia; Ma JH, China; Shao JQ, China; Yuan J, China S-Editor: Wang JJ L-Editor: A P-Editor: Chen YX

References
1.  Galicia-Garcia U, Benito-Vicente A, Jebari S, Larrea-Sebal A, Siddiqi H, Uribe KB, Ostolaza H, Martín C. Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 753]  [Article Influence: 188.3]  [Reference Citation Analysis (0)]
2.  Khan MAB, Hashim MJ, King JK, Govender RD, Mustafa H, Al Kaabi J. Epidemiology of Type 2 Diabetes - Global Burden of Disease and Forecasted Trends. J Epidemiol Glob Health. 2020;10:107-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 1027]  [Article Influence: 342.3]  [Reference Citation Analysis (1)]
3.  Laakso M, Fernandes Silva L. Genetics of Type 2 Diabetes: Past, Present, and Future. Nutrients. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
4.  Kaul N, Ali S. Genes, Genetics, and Environment in Type 2 Diabetes: Implication in Personalized Medicine. DNA Cell Biol. 2016;35:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
5.  Mambiya M, Shang M, Wang Y, Li Q, Liu S, Yang L, Zhang Q, Zhang K, Liu M, Nie F, Zeng F, Liu W. The Play of Genes and Non-genetic Factors on Type 2 Diabetes. Front Public Health. 2019;7:349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
6.  Shoily SS, Ahsan T, Fatema K, Sajib AA. Common genetic variants and pathways in diabetes and associated complications and vulnerability of populations with different ethnic origins. Sci Rep. 2021;11:7504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
7.  Sikhayeva N, Iskakova A, Saigi-Morgui N, Zholdybaeva E, Eap CB, Ramanculov E. Association between 28 single nucleotide polymorphisms and type 2 diabetes mellitus in the Kazakh population: a case-control study. BMC Med Genet. 2017;18:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
8.  Chen M, Zhang X, Fang Q, Wang T, Li T, Qiao H. Three single nucleotide polymorphisms associated with type 2 diabetes mellitus in a Chinese population. Exp Ther Med. 2017;13:121-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Hubacek JA, Dlouha L, Adamkova V, Dlouha D, Pacal L, Kankova K, Galuska D, Lanska V, Veleba J, Pelikanova T. Genetic risk score is associated with T2DM and diabetes complications risks. Gene. 2023;849:146921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
10.  Shitomi-Jones LM, Akam L, Hunter D, Singh P, Mastana S. Genetic Risk Scores for the Determination of Type 2 Diabetes Mellitus (T2DM) in North India. Int J Environ Res Public Health. 2023;20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
11.  Duschek E, Forer L, Schönherr S, Gieger C, Peters A, Kronenberg F, Grallert H, Lamina C. A polygenic and family risk score are both independently associated with risk of type 2 diabetes in a population-based study. Sci Rep. 2023;13:4805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
12.  Ramos-Lopez O, Milagro FI, Allayee H, Chmurzynska A, Choi MS, Curi R, De Caterina R, Ferguson LR, Goni L, Kang JX, Kohlmeier M, Marti A, Moreno LA, Pérusse L, Prasad C, Qi L, Reifen R, Riezu-Boj JI, San-Cristobal R, Santos JL, Martínez JA. Guide for Current Nutrigenetic, Nutrigenomic, and Nutriepigenetic Approaches for Precision Nutrition Involving the Prevention and Management of Chronic Diseases Associated with Obesity. J Nutrigenet Nutrigenomics. 2017;10:43-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 90]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
13.  Ortega Á, Berná G, Rojas A, Martín F, Soria B. Gene-Diet Interactions in Type 2 Diabetes: The Chicken and Egg Debate. Int J Mol Sci. 2017;18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
14.  Dietrich S, Jacobs S, Zheng JS, Meidtner K, Schwingshackl L, Schulze MB. Gene-lifestyle interaction on risk of type 2 diabetes: A systematic review. Obes Rev. 2019;20:1557-1571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
15.  Sorgini C, Christensen J, Parnell L, Tucker K, Ordovas JM, Lai CQ. Predicting Type 2 Diabetes Incidence with Genome-wide Gene-gene and Gene-diet Interactions (OR31-08-19). Curr Dev Nutr. 2019;3:nzz037.OR31-08.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
16.  Ericson U, Hindy G, Drake I, Schulz CA, Brunkwall L, Hellstrand S, Almgren P, Orho-Melander M. Dietary and genetic risk scores and incidence of type 2 diabetes. Genes Nutr. 2018;13:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
17.  Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Cuervo M, Goni L, Martinez JA. Interplay of an Obesity-Based Genetic Risk Score with Dietary and Endocrine Factors on Insulin Resistance. Nutrients. 2019;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
18.  Alsulami S, Cruvinel NT, da Silva NR, Antoneli AC, Lovegrove JA, Horst MA, Vimaleswaran KS. Effect of dietary fat intake and genetic risk on glucose and insulin-related traits in Brazilian young adults. J Diabetes Metab Disord. 2021;20:1337-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
19.  Alathari BE, Nyakotey DA, Bawah AM, Lovegrove JA, Annan RA, Ellahi B, Vimaleswaran KS. Interactions between Vitamin D Genetic Risk and Dietary Factors on Metabolic Disease-Related Outcomes in Ghanaian Adults. Nutrients. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
20.  Hong KW, Kim SH, Zhang X, Park S. Interactions among the variants of insulin-related genes and nutrients increase the risk of type 2 diabetes. Nutr Res. 2018;51:82-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
21.  Choi Y, Kwon HK, Park S. Polygenic Variants Linked to Oxidative Stress and the Antioxidant System Are Associated with Type 2 Diabetes Risk and Interact with Lifestyle Factors. Antioxidants (Basel). 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
22.  Wang W, Jiang H, Zhang Z, Duan W, Han T, Sun C. Interaction between dietary branched-chain amino acids and genetic risk score on the risk of type 2 diabetes in Chinese. Genes Nutr. 2021;16:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
23.  Daily JW, Liu M, Park S. High genetic risk scores of SLIT3, PLEKHA5 and PPP2R2C variants increased insulin resistance and interacted with coffee and caffeine consumption in middle-aged adults. Nutr Metab Cardiovasc Dis. 2019;29:79-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
24.  Yu H, Wang T, Zhang R, Yan J, Jiang F, Li S, Jia W, Hu C. Alcohol consumption and its interaction with genetic variants are strongly associated with the risk of type 2 diabetes: a prospective cohort study. Nutr Metab (Lond). 2019;16:64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
25.  López-Portillo ML, Huidobro A, Tobar-Calfucoy E, Yáñez C, Retamales-Ortega R, Garrido-Tapia M, Acevedo J, Paredes F, Cid-Ossandon V, Ferreccio C, Verdugo RA. The Association between Fasting Glucose and Sugar Sweetened Beverages Intake Is Greater in Latin Americans with a High Polygenic Risk Score for Type 2 Diabetes Mellitus. Nutrients. 2021;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
26.  Jia X, Xuan L, Dai H, Zhu W, Deng C, Wang T, Li M, Zhao Z, Xu Y, Lu J, Bi Y, Wang W, Chen Y, Xu M, Ning G. Fruit intake, genetic risk and type 2 diabetes: a population-based gene-diet interaction analysis. Eur J Nutr. 2021;60:2769-2779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
27.  Alsulami S, Bodhini D, Sudha V, Shanthi Rani CS, Pradeepa R, Anjana RM, Radha V, Lovegrove JA, Gayathri R, Mohan V, Vimaleswaran KS. Lower Dietary Intake of Plant Protein Is Associated with Genetic Risk of Diabetes-Related Traits in Urban Asian Indian Adults. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
28.  Kim DS, Kim BC, Daily JW, Park S. High genetic risk scores for impaired insulin secretory capacity doubles the risk for type 2 diabetes in Asians and is exacerbated by Western-type diets. Diabetes Metab Res Rev. 2018;34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
29.  Hur HJ, Yang HJ, Kim MJ, Lee KH, Kim MS, Park S. Association of Polygenic Variants with Type 2 Diabetes Risk and Their Interaction with Lifestyles in Asians. Nutrients. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
30.  Park S. Association of polygenic risk scores for insulin resistance risk and their interaction with a plant-based diet, especially fruits, vitamin C, and flavonoid intake, in Asian adults. Nutrition. 2023;111:112007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
31.  Khamis AM. Pathophysiology, Diagnostic Criteria, and Approaches to Type 2 Diabetes Remission. Cureus. 2023;15:e33908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
32.  Zepeda-Carrillo EA, Ramos-Lopez O, Martínez-López E, Barrón-Cabrera E, Bernal-Pérez JA, Velasco-González LE, Rangel-Rios E, Bustamante Martínez JF, Torres-Valadez R. Effect of Metformin on Glycemic Control Regarding Carriers of the SLC22A1/OCT1 (rs628031) Polymorphism and Its Interactions with Dietary Micronutrients in Type 2 Diabetes. Diabetes Metab Syndr Obes. 2022;15:1771-1784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
33.  Torres-Valadez R, Ramos-Lopez O, Frías Delgadillo KJ, Flores-García A, Rojas Carrillo E, Aguiar-García P, Bernal Pérez JA, Martinez-Lopez E, Martínez JA, Zepeda-Carrillo EA. Impact of APOE Alleles-by-Diet Interactions on Glycemic and Lipid Features- A Cross-Sectional Study of a Cohort of Type 2 Diabetes Patients from Western Mexico: Implications for Personalized Medicine. Pharmgenomics Pers Med. 2020;13:655-663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
34.  Ramos-Lopez O, Mejia-Godoy R, Frías-Delgadillo KJ, Torres-Valadez R, Flores-García A, Sánchez-Enríquez S, Aguiar-García P, Martínez-López E, Zepeda-Carrillo EA. Interactions between DRD2/ANKK1 TaqIA Polymorphism and Dietary Factors Influence Plasma Triglyceride Concentrations in Diabetic Patients from Western Mexico: A Cross-sectional Study. Nutrients. 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
35.  Naeini Z, Abaj F, Rafiee M, Koohdani F. Interactions of BDNF Val66met and dietary indices in relation to metabolic markers among patient with type 2 diabetes mellitus: a cross-sectional study. J Health Popul Nutr. 2023;42:34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
36.  Abaj F, Esmaeily Z, Naeini Z, Rafiee M, Koohdani F. Dietary acid load modifies the effects of ApoA2-265 T > C polymorphism on lipid profile and serum leptin and ghrelin levels among type 2 diabetic patients. BMC Endocr Disord. 2022;22:190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
37.  Abaj F, Rafiee M, Koohdani F. Interaction between CETP polymorphism and dietary insulin index and load in relation to cardiovascular risk factors in diabetic adults. Sci Rep. 2021;11:15906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
38.  Abaj F, Rafiee M, Koohdani F. A personalised diet approach study: Interaction between PPAR-γ Pro12Ala and dietary insulin indices on metabolic markers in diabetic patients. J Hum Nutr Diet. 2022;35:663-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
39.  Abaj F, Koohdani F. Macronutrient intake modulates impact of EcoRI polymorphism of ApoB gene on lipid profile and inflammatory markers in patients with type 2 diabetes. Sci Rep. 2022;12:10504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
40.  Rafiee M, Sotoudeh G, Djalali M, Alvandi E, Eshraghian M, Javadi F, Doostan F, Koohdani F. The interaction between apolipoprotein B insertion/deletion polymorphism and macronutrient intake on lipid profile and serum leptin and ghrelin levels in type 2 diabetes mellitus patients. Eur J Nutr. 2019;58:1055-1065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
41.  Rafiee M, Sotoudeh G, Djalali M, Alvandi E, Eshraghian M, Sojoudi F, Koohdani F. Dietary ω-3 polyunsaturated fatty acid intake modulates impact of Insertion/Deletion polymorphism of ApoB gene on obesity risk in type 2 diabetic patients. Nutrition. 2016;32:1110-1115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Mutch DM, Lowry DE, Roth M, Sihag J, Hammad SS, Taylor CG, Zahradka P, Connelly PW, West SG, Bowen K, Kris-Etherton PM, Lamarche B, Couture P, Guay V, Jenkins DJA, Eck P, Jones PJH. Polymorphisms in the stearoyl-CoA desaturase gene modify blood glucose response to dietary oils varying in MUFA content in adults with obesity. Br J Nutr. 2022;127:503-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
43.  Goni L, Qi L, Cuervo M, Milagro FI, Saris WH, MacDonald IA, Langin D, Astrup A, Arner P, Oppert JM, Svendstrup M, Blaak EE, Sørensen TI, Hansen T, Martínez JA. Effect of the interaction between diet composition and the PPM1K genetic variant on insulin resistance and β cell function markers during weight loss: results from the Nutrient Gene Interactions in Human Obesity: implications for dietary guidelines (NUGENOB) randomized trial. Am J Clin Nutr. 2017;106:902-908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
44.  Grau K, Cauchi S, Holst C, Astrup A, Martinez JA, Saris WH, Blaak EE, Oppert JM, Arner P, Rössner S, Macdonald IA, Klimcakova E, Langin D, Pedersen O, Froguel P, Sørensen TI. TCF7L2 rs7903146-macronutrient interaction in obese individuals' responses to a 10-wk randomized hypoenergetic diet. Am J Clin Nutr. 2010;91:472-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
45.  Qi Q, Bray GA, Hu FB, Sacks FM, Qi L. Weight-loss diets modify glucose-dependent insulinotropic polypeptide receptor rs2287019 genotype effects on changes in body weight, fasting glucose, and insulin resistance: the Preventing Overweight Using Novel Dietary Strategies trial. Am J Clin Nutr. 2012;95:506-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
46.  Qi Q, Bray GA, Smith SR, Hu FB, Sacks FM, Qi L. Insulin receptor substrate 1 gene variation modifies insulin resistance response to weight-loss diets in a 2-year randomized trial: the Preventing Overweight Using Novel Dietary Strategies (POUNDS LOST) trial. Circulation. 2011;124:563-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
47.  Qi Q, Zheng Y, Huang T, Rood J, Bray GA, Sacks FM, Qi L. Vitamin D metabolism-related genetic variants, dietary protein intake and improvement of insulin resistance in a 2 year weight-loss trial: POUNDS Lost. Diabetologia. 2015;58:2791-2799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
48.  Huang T, Huang J, Qi Q, Li Y, Bray GA, Rood J, Sacks FM, Qi L. PCSK7 genotype modifies effect of a weight-loss diet on 2-year changes of insulin resistance: the POUNDS LOST trial. Diabetes Care. 2015;38:439-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
49.  Zheng Y, Huang T, Zhang X, Rood J, Bray GA, Sacks FM, Qi L. Dietary Fat Modifies the Effects of FTO Genotype on Changes in Insulin Sensitivity. J Nutr. 2015;145:977-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
50.  de Luis DA, Aller R, Izaola O, de la Fuente B, Romero E. GENETIC VARIATION IN THE BETA-3-ADRENORECEPTOR GENE (TRP64ARG POLYMORPHISM) AND THEIR INFLUENCE ON ANTHROPOMETRIC PARAMETERS AND INSULIN RESISTANCE AFTER A HIGH PROTEIN/LOW CARBOHYDRATE VERSUS A STANDARD HYPOCALORIC DIET. Nutr Hosp. 2015;32:487-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
51.  de Luis DA, Romero E, Izaola O, Primo D, Aller R. Cardiovascular Risk Factors and Insulin Resistance after Two Hypocaloric Diets with Different Fat Distribution in Obese Subjects: Effect of the rs10767664 Gene Variant in Brain-Derived Neurotrophic Factor. J Nutrigenet Nutrigenomics. 2017;10:163-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
52.  de Luis DA, Aller R, Izaola O, Gonzalez Sagrado M, Conde R. Role of G308 promoter variant of tumor necrosis factor alpha gene on weight loss and metabolic parameters after a high monounsaturated versus a high polyunsaturated fat hypocaloric diets. Med Clin (Barc). 2013;141:189-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
53.  de Luis DA, Aller R, Izaola O, Romero E. Effect of -55CT Polymorphism of UCP3 on Insulin Resistance and Cardiovascular Risk Factors after a High Protein/Low Carbohydrate versus a Standard Hypocaloric Diet. Ann Nutr Metab. 2016;68:157-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
54.  de Luis DA, Izaola O, Primo D, Gómez-Hoyos E, Ortola A, López-Gómez JJ, Aller R. Role of rs1501299 variant in the adiponectin gene on total adiponectin levels, insulin resistance and weight loss after a Mediterranean hypocaloric diet. Diabetes Res Clin Pract. 2019;148:262-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
55.  de Luis D, Aller R, Izaola O, Primo D. Role of the rs10401670 variant in the resistin gene on the metabolic response after weight loss secondary to a high-fat hypocaloric diet with a Mediterranean pattern. J Hum Nutr Diet. 2022;35:722-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
56.  de Luis DA, Izaola O, Primo D, Aller R. Association of the rs10830963 polymorphism in melatonin receptor type 1B (MTNR1B) with metabolic response after weight loss secondary to a hypocaloric diet based in Mediterranean style. Clin Nutr. 2018;37:1563-1568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
57.  Wang T, Huang T, Zheng Y, Rood J, Bray GA, Sacks FM, Qi L. Genetic variation of fasting glucose and changes in glycemia in response to 2-year weight-loss diet intervention: the POUNDS LOST trial. Int J Obes (Lond). 2016;40:1164-1169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
58.  Huang T, Ley SH, Zheng Y, Wang T, Bray GA, Sacks FM, Qi L. Genetic susceptibility to diabetes and long-term improvement of insulin resistance and β cell function during weight loss: the Preventing Overweight Using Novel Dietary Strategies (POUNDS LOST) trial. Am J Clin Nutr. 2016;104:198-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
59.  Han L, Ma W, Sun D, Heianza Y, Wang T, Zheng Y, Huang T, Duan D, Bray JGA, Champagne CM, Sacks FM, Qi L. Genetic variation of habitual coffee consumption and glycemic changes in response to weight-loss diet intervention: the Preventing Overweight Using Novel Dietary Strategies (POUNDS LOST) trial. Am J Clin Nutr. 2017;106:1321-1326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
60.  Kim M, Chae JS, Kim M, Lee SH, Lee JH. Effects of a 3-year dietary intervention on age-related changes in triglyceride and apolipoprotein A-V levels in patients with impaired fasting glucose or new-onset type 2 diabetes as a function of the APOA5 -1131 T > C polymorphism. Nutr J. 2014;13:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
61.  Liu PJ, Liu YP, Qin HK, Xing T, Li SS, Bao YY. Effects of polymorphism in FABP2 Ala54Thr on serum lipids and glycemic control in low glycemic index diets are associated with gender among Han Chinese with type 2 diabetes mellitus. Diabetes Metab Syndr Obes. 2019;12:413-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
62.  Hosseini S, Alipour M, Zakerkish M, Cheraghian B, Ghandil P. Effects of epigallocatechin gallate on total antioxidant capacity, biomarkers of systemic low-grade inflammation and metabolic risk factors in patients with type 2 diabetes mellitus: the role of FTO-rs9939609 polymorphism. Arch Med Sci. 2021;17:1722-1729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
63.  Vranceanu M, Pickering C, Filip L, Pralea IE, Sundaram S, Al-Saleh A, Popa DS, Grimaldi KA. A comparison of a ketogenic diet with a LowGI/nutrigenetic diet over 6 months for weight loss and 18-month follow-up. BMC Nutr. 2020;6:53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
64.  Ojeda-Granados C, Panduro A, Rivera-Iñiguez I, Sepúlveda-Villegas M, Roman S. A Regionalized Genome-Based Mexican Diet Improves Anthropometric and Metabolic Parameters in Subjects at Risk for Obesity-Related Chronic Diseases. Nutrients. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
65.  Frankwich KA, Egnatios J, Kenyon ML, Rutledge TR, Liao PS, Gupta S, Herbst KL, Zarrinpar A. Differences in Weight Loss Between Persons on Standard Balanced vs Nutrigenetic Diets in a Randomized Controlled Trial. Clin Gastroenterol Hepatol. 2015;13:1625-1632.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
66.  Gkouskou K, Lazou E, Skoufas E, Eliopoulos AG. Genetically Guided Mediterranean Diet for the Personalized Nutritional Management of Type 2 Diabetes Mellitus. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
67.  Gkouskou KK, Grammatikopoulou MG, Lazou E, Sanoudou D, Goulis DG, Eliopoulos AG. Genetically-Guided Medical Nutrition Therapy in Type 2 Diabetes Mellitus and Pre-diabetes: A Series of n-of-1 Superiority Trials. Front Nutr. 2022;9:772243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
68.  Waxler JL, O'Brien KE, Delahanty LM, Meigs JB, Florez JC, Park ER, Pober BR, Grant RW. Genetic counseling as a tool for type 2 diabetes prevention: a genetic counseling framework for common polygenetic disorders. J Genet Couns. 2012;21:684-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
69.  Grant RW, O'Brien KE, Waxler JL, Vassy JL, Delahanty LM, Bissett LG, Green RC, Stember KG, Guiducci C, Park ER, Florez JC, Meigs JB. Personalized genetic risk counseling to motivate diabetes prevention: a randomized trial. Diabetes Care. 2013;36:13-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
70.  Voils CI, Coffman CJ, Grubber JM, Edelman D, Sadeghpour A, Maciejewski ML, Bolton J, Cho A, Ginsburg GS, Yancy WS Jr. Does Type 2 Diabetes Genetic Testing and Counseling Reduce Modifiable Risk Factors? A Randomized Controlled Trial of Veterans. J Gen Intern Med. 2015;30:1591-1598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
71.  Wang S, Yong H, He XD. Multi-omics: Opportunities for research on mechanism of type 2 diabetes mellitus. World J Diabetes. 2021;12:1070-1080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
72.  Raciti GA, Nigro C, Longo M, Parrillo L, Miele C, Formisano P, Béguinot F. Personalized medicine and type 2 diabetes: lesson from epigenetics. Epigenomics. 2014;6:229-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
73.  Qie R, Han M, Huang S, Li Q, Liu L, Zhang D, Cheng C, Zhao Y, Liu D, Qin P, Guo C, Zhou Q, Tian G, Zhang Y, Wu X, Wu Y, Li Y, Yang X, Feng Y, Hu F, Zhang M, Hu D, Lu J. Association of TCF7L2 gene polymorphisms, methylation, and gene-environment interaction with type 2 diabetes mellitus risk: A nested case-control study in the Rural Chinese Cohort Study. J Diabetes Complications. 2021;35:107829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
74.  Gong W, Xiao D, Ming G, Yin J, Zhou H, Liu Z. Type 2 diabetes mellitus-related genetic polymorphisms in microRNAs and microRNA target sites. J Diabetes. 2014;6:279-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
75.  Safari-Alighiarloo N, Emami Z, Rezaei-Tavirani M, Alaei-Shahmiri F, Razavi S. Gut Microbiota and Their Associated Metabolites in Diabetes: A Cross Talk Between Host and Microbes-A Review. Metab Syndr Relat Disord. 2023;21:3-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
76.  Bailén M, Tabone M, Bressa C, Lominchar MGM, Larrosa M, González-Soltero R. Unraveling Gut Microbiota Signatures Associated with PPARD and PARGC1A Genetic Polymorphisms in a Healthy Population. Genes (Basel). 2022;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
77.  Kwan SY, Sabotta CM, Joon A, Wei P, Petty LE, Below JE, Wu X, Zhang J, Jenq RR, Hawk ET, McCormick JB, Fisher-Hoch SP, Beretta L. Gut Microbiome Alterations Associated with Diabetes in Mexican Americans in South Texas. mSystems. 2022;7:e0003322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
78.  Chen ZZ, Gerszten RE. Metabolomics and Proteomics in Type 2 Diabetes. Circ Res. 2020;126:1613-1627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
79.  Ghanbari F, Yazdanpanah N, Yazdanpanah M, Richards JB, Manousaki D. Connecting Genomics and Proteomics to Identify Protein Biomarkers for Adult and Youth-Onset Type 2 Diabetes: A Two-Sample Mendelian Randomization Study. Diabetes. 2022;71:1324-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
80.  Beijer K, Nowak C, Sundström J, Ärnlöv J, Fall T, Lind L. In search of causal pathways in diabetes: a study using proteomics and genotyping data from a cross-sectional study. Diabetologia. 2019;62:1998-2006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
81.  Rohde PD, Nyegaard M, Kjolby M, Sørensen P. Multi-Trait Genomic Risk Stratification for Type 2 Diabetes. Front Med (Lausanne). 2021;8:711208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
82.  Timasheva Y, Balkhiyarova Z, Avzaletdinova D, Rassoleeva I, Morugova TV, Korytina G, Prokopenko I, Kochetova O. Integrating Common Risk Factors with Polygenic Scores Improves the Prediction of Type 2 Diabetes. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
83.  Ouhaibi-Djellouli H, Mediene-Benchekor S, Lardjam-Hetraf SA, Hamani-Medjaoui I, Meroufel DN, Boulenouar H, Hermant X, Saidi-Mehtar N, Amouyel P, Houti L, Goumidi L, Meirhaeghe A. The TCF7L2 rs7903146 polymorphism, dietary intakes and type 2 diabetes risk in an Algerian population. BMC Genet. 2014;15:134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
84.  Hindy G, Sonestedt E, Ericson U, Jing XJ, Zhou Y, Hansson O, Renström E, Wirfält E, Orho-Melander M. Role of TCF7L2 risk variant and dietary fibre intake on incident type 2 diabetes. Diabetologia. 2012;55:2646-2654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
85.  Wirström T, Hilding A, Gu HF, Östenson CG, Björklund A. Consumption of whole grain reduces risk of deteriorating glucose tolerance, including progression to prediabetes. Am J Clin Nutr. 2013;97:179-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
86.  Bauer W, Adamska-Patruno E, Krasowska U, Moroz M, Fiedorczuk J, Czajkowski P, Bielska D, Gorska M, Kretowski A. Dietary Macronutrient Intake May Influence the Effects of TCF7L2 rs7901695 Genetic Variants on Glucose Homeostasis and Obesity-Related Parameters: A Cross-Sectional Population-Based Study. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
87.  Shin D, Lee KW. Dietary carbohydrates interact with AMY1 polymorphisms to influence the incidence of type 2 diabetes in Korean adults. Sci Rep. 2021;11:16788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
88.  Lee KW, Shin D. Interactions between Bitter Taste Receptor Gene Variants and Dietary Intake Are Associated with the Incidence of Type 2 Diabetes Mellitus in Middle-Aged and Older Korean Adults. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
89.  Lamri A, Abi Khalil C, Jaziri R, Velho G, Lantieri O, Vol S, Froguel P, Balkau B, Marre M, Fumeron F. Dietary fat intake and polymorphisms at the PPARG locus modulate BMI and type 2 diabetes risk in the D.E.S.I.R. prospective study. Int J Obes (Lond). 2012;36:218-224.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Choi WJ, Jin HS, Kim SS, Shin D. Dietary Protein and Fat Intake Affects Diabetes Risk with CDKAL1 Genetic Variants in Korean Adults. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
91.  Lee JK, Kim K, Ahn Y, Yang M, Lee JE. Habitual coffee intake, genetic polymorphisms, and type 2 diabetes. Eur J Endocrinol. 2015;172:595-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
92.  Park S, Kim BC, Kang S. Interaction effect of PGC-1α rs10517030 variants and energy intake in the risk of type 2 diabetes in middle-aged adults. Eur J Clin Nutr. 2017;71:1442-1448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
93.  Daily JW, Park S. Interaction of BDNF rs6265 variants and energy and protein intake in the risk for glucose intolerance and type 2 diabetes in middle-aged adults. Nutrition. 2017;33:187-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
94.  Park S, Zhang X, Lee NR, Jin HS. TRPV1 Gene Polymorphisms Are Associated with Type 2 Diabetes by Their Interaction with Fat Consumption in the Korean Genome Epidemiology Study. J Nutrigenet Nutrigenomics. 2016;9:47-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
95.  Lee YJ, Lee H, Jang HB, Yoo MG, Im S, Koo SK, Lee HJ. The potential effects of HECTD4 variants on fasting glucose and triglyceride levels in relation to prevalence of type 2 diabetes based on alcohol intake. Arch Toxicol. 2022;96:2487-2499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
96.  Shen L, Wang Z, Zang J, Liu H, Lu Y, He X, Wu C, Su J, Zhu Z. The Association between Dietary Iron Intake, SNP of the MTNR1B rs10830963, and Glucose Metabolism in Chinese Population. Nutrients. 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
97.  Lopez-Minguez J, Saxena R, Bandín C, Scheer FA, Garaulet M. Late dinner impairs glucose tolerance in MTNR1B risk allele carriers: A randomized, cross-over study. Clin Nutr. 2018;37:1133-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 70]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
98.  Garaulet M, Lopez-Minguez J, Dashti HS, Vetter C, Hernández-Martínez AM, Pérez-Ayala M, Baraza JC, Wang W, Florez JC, Scheer FAJL, Saxena R. Interplay of Dinner Timing and MTNR1B Type 2 Diabetes Risk Variant on Glucose Tolerance and Insulin Secretion: A Randomized Crossover Trial. Diabetes Care. 2022;45:512-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
99.  Ericson U, Rukh G, Stojkovic I, Sonestedt E, Gullberg B, Wirfält E, Wallström P, Orho-Melander M. Sex-specific interactions between the IRS1 polymorphism and intakes of carbohydrates and fat on incident type 2 diabetes. Am J Clin Nutr. 2013;97:208-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
100.  Zheng JS, Arnett DK, Parnell LD, Smith CE, Li D, Borecki IB, Tucker KL, Ordovás JM, Lai CQ. Modulation by dietary fat and carbohydrate of IRS1 association with type 2 diabetes traits in two populations of different ancestries. Diabetes Care. 2013;36:2621-2627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
101.  Sonestedt E, Lyssenko V, Ericson U, Gullberg B, Wirfält E, Groop L, Orho-Melander M. Genetic variation in the glucose-dependent insulinotropic polypeptide receptor modifies the association between carbohydrate and fat intake and risk of type 2 diabetes in the Malmo Diet and Cancer cohort. J Clin Endocrinol Metab. 2012;97:E810-E818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
102.  Blanco-Rojo R, Delgado-Lista J, Lee YC, Lai CQ, Perez-Martinez P, Rangel-Zuñiga O, Smith CE, Hidalgo B, Alcala-Diaz JF, Gomez-Delgado F, Parnell LD, Arnett DK, Tucker KL, Lopez-Miranda J, Ordovas JM. Interaction of an S100A9 gene variant with saturated fat and carbohydrates to modulate insulin resistance in 3 populations of different ancestries. Am J Clin Nutr. 2016;104:508-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
103.  Zheng JS, Arnett DK, Parnell LD, Lee YC, Ma Y, Smith CE, Richardson K, Li D, Borecki IB, Ordovas JM, Tucker KL, Lai CQ. Genetic variants at PSMD3 interact with dietary fat and carbohydrate to modulate insulin resistance. J Nutr. 2013;143:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
104.  Zheng JS, Arnett DK, Parnell LD, Lee YC, Ma Y, Smith CE, Richardson K, Li D, Borecki IB, Tucker KL, Ordovás JM, Lai CQ. Polyunsaturated Fatty Acids Modulate the Association between PIK3CA-KCNMB3 Genetic Variants and Insulin Resistance. PLoS One. 2013;8:e67394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
105.  Fisher E, Schreiber S, Joost HG, Boeing H, Döring F. A two-step association study identifies CAV2 rs2270188 single nucleotide polymorphism interaction with fat intake in type 2 diabetes risk. J Nutr. 2011;141:177-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
106.  Hindy G, Mollet IG, Rukh G, Ericson U, Orho-Melander M. Several type 2 diabetes-associated variants in genes annotated to WNT signaling interact with dietary fiber in relation to incidence of type 2 diabetes. Genes Nutr. 2016;11:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
107.  Nishiya Y, Daimon M, Mizushiri S, Murakami H, Tanabe J, Matsuhashi Y, Yanagimachi M, Tokuda I, Sawada K, Ihara K. Nutrient consumption-dependent association of a glucagon-like peptide-1 receptor gene polymorphism with insulin secretion. Sci Rep. 2020;10:16382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
108.  Ortega-Azorín C, Sorlí JV, Asensio EM, Coltell O, Martínez-González MÁ, Salas-Salvadó J, Covas MI, Arós F, Lapetra J, Serra-Majem L, Gómez-Gracia E, Fiol M, Sáez-Tormo G, Pintó X, Muñoz MA, Ros E, Ordovás JM, Estruch R, Corella D. Associations of the FTO rs9939609 and the MC4R rs17782313 polymorphisms with type 2 diabetes are modulated by diet, being higher when adherence to the Mediterranean diet pattern is low. Cardiovasc Diabetol. 2012;11:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 108]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]