Skip to main content
Physiology logoLink to Physiology
. 2022 Sep 20;38(1):42–50. doi: 10.1152/physiol.00014.2022

Precision Nutrition: Recent Advances in Obesity

V Saroja Voruganti 1,
PMCID: PMC9705019  PMID: 36125787

Abstract

“Precision nutrition” is an emerging area of nutrition research that focuses on understanding metabolic variability within and between individuals and helps develop customized dietary plans and interventions to maintain optimal individual health. It encompasses nutritional genomic (gene-nutrient interactions), epigenetic, microbiome, and environmental factors. Obesity is a complex disease that is affected by genetic and environmental factors and thus a relevant target of precision nutrition-based approaches. Recent studies have shown significant associations between obesity phenotypes (body weight, body mass index, waist circumference, and central and regional adiposity) and genetic variants, epigenetic factors (DNA methylation and noncoding RNA), microbial species, and environment (sociodemographics and physical activity). Additionally, studies have also shown that the interactions between genetic variants, microbial metabolites, and epigenetic factors affect energy balance and adiposity. These include variants in FTO, MC4R, PPAR, APOA, and FADS genes, DNA methylation in CpG island regions, and specific miRNAs and microbial species such as Firmicutes, Bacteriodes, Clostridiales, etc. Similarly, studies have shown that microbial metabolites, folate, B-vitamins, and short-chain fatty acids interact with miRNAs to influence obesity phenotypes. With the advent of next-generation sequencing and analytical approaches, the advances in precision nutrition have the potential to lead to new paradigms, which can further lead to interventions or customized treatments specific to individuals or susceptible groups of individuals. This review highlights the recent advances in precision nutrition as applied to obesity and projects the importance of precision nutrition in obesity and weight management.

Keywords: epigenetics, metabolic individuality, metabolomics, microbiome, nutrigenomics

Introduction

People have different responses to and requirements for nutrients and other components of the diet, and this heterogeneity is an important source of variance in nutrition studies. Understanding this variation and its role in risk for diseases has gained more momentum with the advent of new tools and technologies (13). “Precision nutrition” is an emerging area of nutrition research that focuses on understanding metabolic variability within and between individuals and helps to develop customized dietary plans and interventions to maintain optimum individual health. It assumes that each person will respond differently and that their energy and dietary needs are different from each other (3, 4). Using one’s DNA (genetics) to predict their response to nutrient intake is only one of the various facets of precision nutrition. Other aspects that influence the variation in individual response to nutrients include epigenetic, microbiome, and environmental factors (1, 4, 5).

The US National Institutes of Health (NIH) has recognized and identified precision nutrition as a key area of research in the pursuit of optimal health (6). As an ancillary study to their “All of Us” study, the NIH made a substantial investment in precision nutrition research, in recognition that one-size-does-not-fit all with respect to dietary intake, metabolism, and disease risk (101). Precision nutrition can be categorized into personalized or individualized nutrition and stratified nutrition, the former being at an individual level whereas the latter one focusing on groups of susceptible individuals (4, 7). Precision nutrition is innovative in that it considers individual differences in people’s genes, environment, and lifestyles. This information is key for targeting treatments for specific group of individuals. Advances in precision nutrition have led to, and continue to lead to, new discoveries that can provide the foundations of interventions or treatments tailored to specific individuals (7, 8).

Obesity is a major health concern worldwide (911). It poses a considerable burden health-wise and cost-wise. It is also a precursor to various other metabolic diseases such as type 2 diabetes, cardiovascular diseases, metabolic syndrome, neurodegenerative diseases, and some types of cancers (12, 13). As per CDC 2017–2018 statistics, 42.4% and 9.2% of adults in the United States have obesity and severe obesity, respectively (102). The regulation of weight and metabolism including the physiological processes has been studied extensively. While the pathogenesis of obesity seems as simple as the difference between ingested and expended calories, scientific evidence shows that obesity is a complex disease and involves genetic, epigenetic, microbiome, psychosocial, and environmental factors, as well as the interactions between them. Additionally, a person’s social, cultural, and behavioral factors also impact a person’s weight and body composition (13, 14).

A key feature of obesity is the dysregulation of energy metabolism. The central nervous system plays a vital role in appetite, satiety, and energy regulation, with the hypothalamus as the master regulator. The arcuate nucleus (ARC), a part of the hypothalamus, is a vital center for the regulation of metabolism and feeding (1517). It contains neurons that express two distinct, functionally antagonistic, peptides; orexigenic [appetite stimulating: neuropeptide Y (NPY) and agouti-related peptide (AgRP)] and anorexigenic [appetite suppressing: pro-opiomelanocortin (POMC) and cocaine and amphetamine-regulated transcript (CART)] peptides. The neuronal signals from the lateral hypothalamus (increase food intake) and parabrachial nucleus (decrease food intake) also contribute to the regulation of appetite (18, 19). The peripheral signals include hormonal signals such as leptin, insulin, and gut hormones [cholecystokinin (CCK), ghrelin, peptide YY (PYY), glucagon-like peptide1 (GLP1), oxyntomodulin, obestatin, and nesfatin] (19, 20). The integration of hormonal and nutritional signals from the circulation and peripheral and central neuronal (including astrocyte-neuron and microglia-neuron and gut-brain axis) signals happens in the ARC to generate a feeding response (1517). Both components of energy metabolism, energy intake and expenditure, have been shown to be altered or modified by genetic variants and/or epigenetic modifications, microbial species and metabolites, lifestyle, and social and psychological factors, with serious implications for obesity (FIGURE 1) (1517). This narrative review highlights and discusses the recent advances in the components of precision nutrition in relation to obesity.

FIGURE 1.

FIGURE 1.

Individuality in metabolic response to diet/nutrition intake Components of energy metabolism, energy intake and expenditure, have been shown to be altered or modified by genetic variants and/or epigenetic modifications, microbial species and metabolites, lifestyle, and social and psychological factors, with serious implications for obesity.

Nutritional Genomics and Obesity

Nutritional genomics (NGx) is the study of how an individual’s genetics affect the way nutrients are metabolized (nutrigenetics) and how nutrients affect the way genes are expressed (nutrigenomics) (8, 21). The main aims of NGx are to identify genetic variants that are associated with diet-related diseases, to underlie the variability in responses to diet or nutrients, and to help develop nutritional strategies that can treat or prevent diseases (3, 8, 21). Among the metabolic diseases, obesity is the most heavily studied within the NGx paradigm. It is well known that lifestyle factors such as diet and exercise play a key role in the development and treatment of obesity and related comorbidities. However, it is also known that weight loss interventions, whether dietary plans or exercise regimens, do not elicit the same response from all individuals. Additionally, the equation of energy in and energy out is not as simple as it seems. The interindividual variability in response to diet or exercise is quite substantial, even after correcting for differences in age, gender, race/ethnicity, and sociodemographic characteristics (3, 22, 23).

The imbalance between energy intake and energy expenditure is a key determinant of obesity. Energy intake occurs through intake of macronutrients, carbohydrates, proteins, and fats. The amount and type of these macronutrients determine adiposity and energy equilibrium. A dynamic time-series analysis of three decades of US data and a decade of data from 164 other countries, after adjustment for total calories, has shown that carbohydrate, and not fat, consumption was associated with short- and long-term increases in weight (24). Meta-analysis of studies of low-carbohydrate and low-fat diets for weight loss showed that, irrespective of total calories and weight loss, reduction in carbohydrates may prove beneficial for improving metabolic syndrome markers, including obesity (2527).

It is also known that simple or added sugars and saturated fatty acids can adversely affect adiposity. Simple sugars, such as glucose, fructose, lactose, and sucrose, are metabolized differently than complex carbohydrates. As per the American Heart Association, the recommended total sugar consumption for US adults is 150 and 100 calories per day for men and women, respectively (28). Although much decreased, the consumption of simple sugars and saturated fatty acid remains higher than the recommended intake. Both the monosaccharides glucose and fructose get metabolized and yield great amounts of fat in the liver. However, they differ in regional adipose deposition with glucose promoting subcutaneous adipose deposition while fructose favors visceral adipose tissue deposition (29). Other studies have shown the adverse effects of simple sugars, either fructose or glucose, on adiposity and weight gain (3033). However, in a recent study investigating the association between macronutrient intakes and obesity and metabolic risk, the authors did not find any link between carbohydrates, fats, and weight (34). These studies and results show differences in the way nutrients affect obesity phenotypes and point to the involvement of other biological and environmental factors.

Individual variability in weight phenotypes has a strong genetic basis. Twin, family-based, and other studies have estimated heritabilities of body composition and related phenotypes to be 40–70% (3538). Genome-wide association and candidate gene studies have identified several genetic variants that affect various components of obesity. In a study of more than 300,000 European adults, Locke et al. (39) identified 97 loci associated with body mass index (BMI). Many of these loci, particularly a specific set of 32 loci, have been consistently reported to be associated with obesity across ethnicities. Some studies have generated polygenic risk scores (PRS) from these 32 loci and found stronger effects on anthropometric measures (40). The PRS is computed from an independent set of polymorphisms that are associated with the risk for a disease. The total number of risk alleles per individual is summed, weighted or unweighted by its effect size, and a score is calculated. A single score, the PRS, thus reflects the cumulative effect of the included polymorphisms and is a better predictor of the disease risk (41, 42). A study in the UK Biobank has generated a PRS with 2.1 million single nucleotide polymorphisms (SNPs) associated with obesity and assessed them in individuals ranging from birth to middle age (43).

Prominent among the genes that are associated with appetite or adiposity are fat mass and obesity related (FTO), melanocortin 4 receptor 4 (MC4R), apolipoprotein A5 (APOA5), neuropeptide Y (NPY), leptin (LEP), leptin receptor (LEPR), brain-derived neurotrophic factor (BDNF), single-minded homolog1 (SIM1), neurexin3 (NRXN3), NPC intracellular cholesterol transporter 1 (NPC1), neuronal growth regulator q (NEGR1), mitochondrial carrier homolog 2 (MTCH2), apolipoprotein E (APOE), tumor necrosis factor alpha (TNFα), plasminogen activator inhibitor 1 (PAI-1), transmembrane protein 18 (TMEM18), etc. (4346). A study investigating genetic influence on extreme obesity found several loci that are linked to either the circadian rhythm of food consumption or hypothalamic signaling related to food intake (47, 48).

Studies have also shown differential effects of nutrients based on an individual’s genetic makeup. Genetic variants interact with nutrition and other environmental factors such as physical activity to affect obesity phenotypes. The most frequently studied nutrients have been simple sugars and saturated and unsaturated fatty acids. Simple sugars such as fructose and glucose, either in the form of sugar-sweetened beverages (SSB) or as part of high fructose/glucose diets, have been studied to understand their effects on obesity and related comorbidities (49, 50). In circulation, fructose and glucose have different metabolic fates, although fructose needs glucose for its metabolic effects. Fructose, more than glucose, favors hepatic lipogenesis (49). A large study including 6,934 women from the Nurses Health Study, 4,423 men from the Health Professionals Follow-up Study, and 21,740 women from the Women’s Genome Health Study generated a PRS, based on the aforementioned 32 obesity-associated SNPs. They found that with an increment per 10 risk alleles, the relative risk for BMI increased with every category of increasing SSB intake (49). Some other examples of gene by diet interactions with respect to obesity include carbohydrate intake and MC4R rs17782313 (51), low-protein diet and genetic risk score (GRS) of 15 SNPs (52), saturated fat intake and FTO (53), vitamin C intake and carboxy peptidase (CP) rs59465035 (54), fat and sugar intake and taste receptor (TAS1R2) (55), 5 SNP PRS and fried foods (56), high-fat hypocaloric diet and resistin (RETN) rs10401670 (57), etc. (Table 1).

TABLE 1.

Summary of nutrigenetics of obesity phenotypes

Variant of Interest Gene Type of Study Sample Size Nutrient/Dietary Intake Outcome Population Conclusion Reference(s)
rs9939609 FTO Cohort 339,000 Total fat BMI European and Hispanic adults TT allele of both SNPs associated with higher BMI with higher SFA intake Corella et al. (53)
rs1121980
rs9939609 FTO Intervention 86 Mediterranean diet Weight loss Italian A allele was associated with less weight loss than TT carriers Franzago et al. (58)
rs662799 APOA5 Cohort 3266 Red meat Metabolic syndrome Korean G carriers and in highest tertile of red meat consumption had 1.7 HR as compared to those with A allele Choi and Shin, (103)
GRS of 4 SNPs Cohort 302 Total fat, SFA, MUFA, and PUFA Waist circumference Ghana GRS >3 and high intake of fat, SFA, PUFA, and MUFA linked to increased waist circumference Alsulami et al. (52)
Fiber Body fat GRS >3 and high intake of fiber had lower total body fat as compared to low fiber intake
GRS of 94 SNPs Cohort 362,496 Alcohol BMI UK adults (UK Biobank) Increase in BMI per GRS is higher in infrequent drinkers as compared to frequent drinkers Rask-Anderson et al. (104)
rs6722579 CAB39 Cohort 50,808 Fat intake Abdominal obesity Korea A carriers were at higher risk for abdominal obesity in those who had fat intake >DRI Kwon et al. (54)
rs59465035 CPQT T carriers are at lower risk for abdominal obesity in those who had vitamin C >DRI
rs17782313 MC4R Cohort 282 Carbohydrate intake BMI Iranian Higher carbohydrate intake was associated with higher BMI and waist circumference in C carriers Alizadeh et al. (51)
rs1801282 PPARG Intervention 327 Diet therapy vs. diet therapy + metformin Weight loss CC carriers had a greater weight loss than others Valeeva et al. (48)
rs59465035 CP Cohort 50,808 Vitamin C Waist circumference Korean T-allele carriers had lower risk of abdominal obesity as compared to others in those who consumed vitamin C higher than DRI Kwon et al. (54)
rs10401670 RETN Intervention 284 High-fat hypocaloric diet BMI Spain T-allele carriers had greater weight loss than others De Luis et al. (57)
5 SNP PRS Cohort >53,000 Fried foods BMI Korean High PRS and menarche linked with higher BMI Park et al. (56)
rs1761667, CD36 Cohort 142 Fat and sugar intake BMI percentiles African American females AA and altered fat perception linked to BMI increase Primeaux et al. (55)
rs35874116 TAS1R2 TT and sugar intake linked to BMI increase
rs713598 TAS2R38 CC and energy intake linked to BMI increase

GRS, genetic risk score; PRS, polygenic risk score; BMI, body mass index; SNP, single nucleotide polymorphism; SFA, saturated fatty acid; MUFA, monounsaturated fatty acids; PUFA, polyunsaturated fatty acids; DRI, dietary reference intake.

Weight loss interventions have also shown better results when participant genetics were considered. For example, a randomized controlled clinical trial, The Nutrigenomics, Overweight/Obesity and Weight Management (NOW) study conducted a weight loss trial with two groups, one with a lifestyle balance program and the other one with a lifestyle balance program with genetic testing. The latter group showed better weight loss than the former one for as long as 6 months (59). Similar results were shown in another trial that had 284 individuals who underwent a high-fat, hypocaloric diet. They found greater weight loss in T carriers of rs10401670 of RETN as compared to others (57). Studies have also shown the effect of a few genetic variants on the extent of weight loss in intervention studies. A study by Franzago et al. (58) found that A-allele carriers of rs9939609 of the FTO gene lost less weight and decreased BMI less than TT carriers during a 12-mo diet/lifestyle intervention. A study by Garaulet et al. (60) found that eating late was linked to lower weight loss in AA genotype of PLIN1 14995 A > T carriers, thus pointing to the importance of meal timing along with genes in obesity. On the other hand, a 10-wk weight-loss intervention based on 35 SNPs did not find any difference between personalized dietary intervention based on 35 SNPs and a generic healthy diet with respect to either loss of body weight or fat mass (61).

In a similar vein, modified nutrient diets have been linked to changes in the expression of genes associated with adiposity. In a collaborative cross mice model, Yam et al (62) showed that genetic background and macronutrient alterations affected the expression of genes involved in adiposity. There is an increasing interest in intermittent fasting and its effect on metabolic diseases including obesity. A study of individuals with overweight/obesity showed a significant decrease in FTO gene expression after the month-long fasting period of Ramadan in contrast to normal weight individuals (63).

Nutritional Epigenetics and Obesity

Epigenetics refer to the genetic effects that are external to changes in DNA sequence. The various types of epigenetic changes include DNA methylation (addition of methyl groups to cysteine or adenine on the DNA), noncoding RNAs (short and long noncoding RNAs that modulate mRNAs to affect gene expression), and histone modification (posttranslational modification of histone proteins that can affect chromatin function and DNA accessibility) (6466). A branch of epigenetics, nutritional epigenetics, is the study of the effects of food on the expression of genes (67). Human and animal studies have shown that epigenetic effects can transcend generations. Maternal nutrition, in particular, can have distinct epigenetic effects on the child’s risk for various metabolic diseases (6870).

An individual’s alleles are determined by their maternal or paternal origins (genomic imprinting), and these are involved in a variety of metabolic functions. Alterations in genomic imprinting due to epigenetic mechanisms such as DNA methylation, histone modifications, or genetic events such as translocation, inversion, duplication, etc., can result in obesity (67). Animal studies have shown that paternal BMI at the time of conception can influence an infant’s birth weight and patterns of DNA methylation in infant cord blood (71). Several studies have evaluated the Dutch Famine Birth Cohort of the Hunger Winter. These studies showed that intrauterine exposure to maternal undernutrition results in long-term effects on a child’s metabolic health, including obesity, type 2 diabetes, and metabolic syndrome (72, 73). Interestingly, these exposures had long-term transgenerational effects. Similar results were shown in studies using data from the Chinese Famine that occurred between 1959 and 1961. Individuals born during this period reported higher cases of obesity, type 2 diabetes, and metabolic syndrome as compared to those born after the famine (74, 75). In another human study, DNA collected from infants born at term with 40 women of normal BMI and with obesity revealed genomic loci associated with differentially methylated regions (DMRs) in CpG-dense regions (70). When compared with another larger study, there was a significant overlap in DMRs, with key genes being PTPRN2, a gene involved in insulin secretion, and MAD1L1, a gene involved in cell cycle and tumor progression (70).

An intervention study that included 12 months of lifestyle modification (Mediterranean diet and physical activity) showed links between BMI and DNA methylation patterns. DNA methylation patterns in genes related to lipid metabolism and inflammation were the most modified by lifestyle interventions (65). In yet another intervention study that involved a very-low-calorie diet (VLCD), the investigators found that after a VLCD therapy in individuals with obesity, DNA methylation patterns changed and resembled more those of a normal weight person (76). In a cohort of normal-weight and overweight-obese individuals, higher BMI was associated with lower methylenetetrahydrofolate reductase (MTHFR) gene methylation (77). Obesity-associated alterations in methylation were also observed in individuals on either weight-loss diets or surgeries. In a group of individuals undergoing either a very-low-calorie ketogenic diet (VLCKD), a balanced hypocaloric diet (BHD), or bariatric surgery, DNA methylation in the angiotensin-converting enzyme (ACE2) was higher in individuals with obesity as compared to normal-weight individuals. VLCKD and BHDs helped reverse this hypermethylation, but bariatric surgery did not show a similar benefit (78).

Noncoding RNA, especially microRNAs (miRNA), is another class of epigenetic markers that have been linked to obesity and risk for other metabolic diseases (79). These miRNA can affect mRNA stability and degradation by binding anywhere along the length of the mRNA transcript (80, 81). Like genetic variants, several loci that correspond to miRNAs have been identified. Kunej et al. (37) identified 1,736 genomic loci associated with obesity, of which 221 correspond to miRNAs. Several studies have also found these miRNA to be correlated with diet and lifestyle (82, 83), key ones being miR-17/20/93, 21-590-5p, 200 b/c, 221/222, let-7/miR-98, and miR-203 families of miRNAs. In a children study, sex-specific association with obesity was found with miRNAs, 26 b-3p, hsa-576-5p, hsa-31-5p, hsa-10b-5p, and hsa-31-5p. (82). The understanding of the human epigenome and its interaction with environment, particularly diet, may provide greater insight into the pathogenesis of obesity and help develop personalized nutritional interventions.

Microbiome and Obesity

The human body contains millions of microbes that can have significant health effects. The microorganisms, collectively termed the microbiome, are from bacterial, fungal, and protozoal origins (84). The composition of gut microbiota is influenced, to a greater extent, by indigestible foods. The microbiota residing in GI tracts prefer fermentable carbohydrates as substrates whereas they use proteins and amino acids for their own enzymes. The distant colon thus gains energy from fermented residual peptides and proteins. The microorganisms residing in the gut have been associated with obesity risk (85, 86). Individuals with obesity have specific alterations in the composition and function of the gut microbiome (8688), and the gut microbiome affects energy balance by affecting both energy utilization from the diet as well as energy expenditures and storage. The effects of gut microbiome on obesity phenotype and weight loss have been captured through population-based cohort studies as well as randomized controlled trials (89, 90).

Studies have shown that individuals with obesity have a greater amount of Firmicutes and higher Firmicutes to Bacteriodes ratios (91). Other studies have shown Firmicutes and Bacteriodes to be inversely and positively associated with energy expenditure and percent body fat, respectively (63). A recent study by Depommier et al. (92) showed that pasteurized Akkermansia muciniphila increased energy expenditure and reduced body weight and fat mass in diet-induced obesity in mice models. The same groups showed that Akkermansia also improved gut barrier integrity, insulin resistance, and dyslipidemia in mice. They also translated these findings to humans in an exploratory study where they found a reduction in markers of liver dysfunction and inflammation after supplementing the diet with Akkermansia (93). Similar results have been shown in humans using other forms of bacteria (88, 90, 93). In a study in normal weight individuals with high and low visceral fat, the investigators found that a total of 16 species of microbes were significantly correlated with visceral fat accumulation as measured by quantitative computed tomography but not with BMI or waist circumference (90). Of these Bacteriodes were associated with low visceral fat. A longitudinal study of obesity conducted in individuals undergoing laparoscopic sleeve gastrectomy (LSG), found a negative correlation between visceral fat and few microbial species, the strongest being with Eubacterium eligens. Before and after LSG analysis showed a significant increase in specific species such as C. symbiosum, C. hathewayi, C. citroniae, and other Clostridiales after LSG (88). This is significant as Clostridiales are considered to be “good” microbes and are negatively associated with metabolic diseases (94).

The gut microbiota, whose genome is more than 100 times the human genome, are also known to pass from the mother to the neonate through exposure during delivery and through the breastfeeding (95). In addition, the microbial metabolites such as short-chain fatty acids, trimethylamine-N-oxide (TMAO), folate, and other B vitamins, etc., are known to modulate epigenetic mechanisms including DNA methylation, histone modifications, and noncoding RNAs (96, 97). The microbiota is increasingly being recognized as a potential synthesizer of biological compounds that can be used as epigenetic substrates or regulators of epigenetic enzyme activity (98).

Conclusions

Precision nutrition is critical to formulating effective dietary plans customized for individuals or a group of susceptible individuals of diverse ethnicities and populations. To develop personalized or stratified dietary plans, either to maintain optimal health or to treat metabolic diseases, there is a need to understand how an individual responds to diet or nutrients and the factors responsible for this variation in response. These areas of nutrition fall under the purview of precision nutrition. Knowledge of genetic variants affecting nutrient metabolic pathways is not new but dates to the recognition of phenylketonuria, lactase persistence, glucose phosphate dehydrogenase deficiency, etc. What is new is that with the advent of next-generation sequencing technologies and the latest analytical approaches, we are now able to conduct a comprehensive investigation using ‘Omics’ approaches, which are revolutionizing the field. However, there are some issues that need to be resolved before precision nutrition can be put into practice. First, self-reporting approaches continue to be the primary methods for collecting dietary data. Unless there is an objective approach to measuring dietary intake, accurate estimates of dietary intake (especially long term) will be difficult. Second, there is a need to better integrate multiple sources of data. Usage of machine learning approaches is helping address the integration of multiple sources of data. Third, proper flow of data among researchers, practitioners, and educators is needed for dissemination or translation of these results. For example, physicians and dietitian need proper training to be able to prescribe genotype-guided diet plans to patients. Fourth, proper counseling of patients is needed for them to be able to comply with the personalized diet. Despite all these caveats, incorporation of precision nutrition approaches has started showing noteworthy results.

In a study conducted by Connell et al. (99), researchers provided participants with personalized data-driven dietary recommendations, using artificial intelligence, and found significant improvement in clinical outcomes in all conditions and disease activities. Using dietary, genetic, and epigenetic data and machine learning approaches, Lee et al. (100) identified 21 SNPs, 230 DNA methylation sites in lipid and carbohydrate metabolism-related genes, and 26 dietary factors that predicted obesity with 70% accuracy. The main goal of precision nutrition is to understand the differences in the metabolic responses to diet/nutrient intake between individuals and develop tailored or customized nutritional plans for optimal health. With the latest Omics and machine-learning-based approaches, the future of precision nutrition, individualized or stratified, is bright and can be an effective adjunct therapy to existing treatment strategies.

Acknowledgments

V. S. Voruganti is funded by the National Institute of Diabetes and Digestive and Kidney Diseases Grants R01DK126666 and P30DK056350, and Eunice Kennedy Shriver National Institute of Child Health and Human Development Grant UG1HD107692.

No conflicts of interest, financial or otherwise, are declared by the authors.

V.S.V. conceived and designed research; V.S.V. drafted manuscript; V.S.V. edited and revised manuscript; V.S.V. approved final version of manuscript.

References

  • 1. Toro-Martín J, Arsenault B, Després JP, Vohl MC. Precision Nutrition: a review of personalized nutritional approaches for the prevention and management of Metabolic Syndrome. Nutrient 9: 913, 2017. doi: 10.3390/nu9080913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Zeisel SH. A conceptual framework for studying and investing in precision nutrition. Front Genet 10: 200, 2019. doi: 10.3389/fgene.2019.00200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Berry SE, Valdes AM, Drew DA, Asnicar F, Mazidi M, Wolf J, Capdevila J, Hadjigeorgiou G, Davies R, Al Khatib H, Bonnett C, Ganesh S, Bakker E, Hart D, Mangino M, Merino J, Linenberg I, Wyatt P, Ordovas JM, Gardner CD, Delahanty LM, Chan AT, Segata N, Franks PW, Spector TD. Human postprandial responses to food and potential for precision nutrition. Nat Med 26: 964–973, 2020. doi: 10.1038/s41591-020-0934-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Zeisel SH. Precision (Personalized) nutrition: understanding metabolic heterogeneity. Annu Rev Food Sci Technol 11: 71–92, 2020. doi: 10.1146/annurev-food-032519-051736. [DOI] [PubMed] [Google Scholar]
  • 5. Kirk D, Catal C, Tekinerdogan B. Precision Nutrition: a systematic literature review. Comput Biol Med 133: 104365, 2021. doi: 10.1016/j.compbiomed.2021.104365. [DOI] [PubMed] [Google Scholar]
  • 6. Rodgers GP, Collins FS. Precision Nutrition–the answer to “what to eat to stay healthy”. JAMA 324: 735–736, 2020. doi: 10.1001/jama.2020.13601. [DOI] [PubMed] [Google Scholar]
  • 7. Mullins VA, Bresette W, Johnstone L, Hallmark B, Chilton FH. Genomics in personalized nutrition: can you “eat for your genes”? Nutrients 12: 3118, 2020. doi: 10.3390/nu12103118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Marcum JA. Nutrigenetics/nutrigenomics, personalized nutrition and precision healthcare. Curr Nutr Rep 9: 338–345, 2020. doi: 10.1007/s13668-020-00327-z. [DOI] [PubMed] [Google Scholar]
  • 9. Bluher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol 15: 288–298, 2019. doi: 10.1038/s41574-019-0176-8. [DOI] [PubMed] [Google Scholar]
  • 10. Safaei M, Sundararajan EA, Driss M, Boulila W, Shapi'i A. A systematic literature review on obesity: undertstanding the causes and consequences of obesity and reviewing various machine learning approaches used to predict obesity. Comp Biol Med 136: 104754, 2021. doi: 10.1016/j.compbiomed.2021.104754. [DOI] [PubMed] [Google Scholar]
  • 11. Gjermeni E, Kirstein AS, Kolbig F, Kirchhof M, Bundalian L, Katzmann JL, Laufs U, Bluher M, Garten A, Le Duc D. Obesity-an update on the basic pathophysiology and review of recent therapeutic advances. Biomolecules 11: 1426, 2021. doi: 10.3390/biom11101426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Herrera BM, Keildson S, Lindgren CM. Genetics and epigenetics of obesity. Maturitas 69: 41–49, 2011. doi: 10.1016/j.maturitas.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Rohde K, Keller M, la Cour Poulsen L, Bluher M, Kovacs P, Bottcher Y. Genetics and epigenetics of obesity. Metabolism 92: 37–50, 2019. doi: 10.1016/j.metabol.2018.10.007. [DOI] [PubMed] [Google Scholar]
  • 14. Bouchard C. Genetics of obesity: what have we learned over decades of research. Obesity (Silver Spring) 29: 802–820, 2021. doi: 10.1002/oby.23116. [DOI] [PubMed] [Google Scholar]
  • 15. Timper K, Bruning JC. Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis Model Mech 10: 679–689, 2017. doi: 10.1242/dmm.026609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Seong J, Kang JY, Sun JS, Kim KW. Hypothalamic inflammation and obesity: a mechanistic review. Arch Pharm Res 42: 383–392, 2019. doi: 10.1007/s12272-019-01138-9. [DOI] [PubMed] [Google Scholar]
  • 17. Rodriguez-Rodriguez R, Miralpeix C. Hypothalamic regulation of obesity. Int J Mol Sci 22: 13459, 2021. doi: 10.3390/ijms222413459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Molfino A, Imbimbo G. Editorial: appetite control in obesity. Front Nutr 9: 959627, 2022. doi: 10.3389/fnut.2022.959627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Miller GD. Appetite regulation: hormones, peptides and neurotransmitters and their role in obesity. Am J Lifestyle Med 13: 586–601, 2019. doi: 10.1177/1559827617716376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Myers MG Jr, Olson DP. Central nervous system control of metabolism. Nature 491: 357–363, 2012. doi: 10.1038/nature11705. [DOI] [PubMed] [Google Scholar]
  • 21. Doo M, Kim Y. Obesity: interactions of genome and nutrients intake. Prev Nutr Food Sci 20: 1–7, 2015. doi: 10.3746/pnf.2015.20.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hattis D, Banati P, Goble R, Burmaster DE. Human interindividual variability in parameters related to health risks. Risk Anal 19: 711–726, 1999. doi: 10.1111/j.1539-6924.1999.tb00441.x. [DOI] [PubMed] [Google Scholar]
  • 23. Barber-Chaomoux N, Milenkovic D, Verny MA, Habauzit V, Pereira B, Lambert C, Richard D, Boby C, Mazur A, Lusson JR, Dobray C, Morand C. Substantial variability across individuals in the vascular and nutrigenomic response to an acute intake of curcumin: a randomized controlled trial. Mol Nutr Food Res 62: 1700418, 2018. doi: 10.1002/mnfr.201700418. [DOI] [PubMed] [Google Scholar]
  • 24. Reira-Crichton D, Teft N. Macronutrients and obesity: revisiting the calories in, calories out framework. Econ Hum Biol 14: 33–49, 2014. doi: 10.1016/j.ehb.2014.04.002. [DOI] [PubMed] [Google Scholar]
  • 25. San-Cristobal R, Navas-Carretero S, Martinez-Gonzalez MA, Ordovas JM, Martinez JA. Contribution of macronutrients to obesity: implications for precision nutrition. Nat Rev Endocrinol 16: 305–320, 2020. doi: 10.1038/s41574-020-0346-8. [DOI] [PubMed] [Google Scholar]
  • 26. Willems AE, Jong MS, van Beek AP, Nederhof E, van Dijk G. Effects of macronutrient intake in obesity: a meta-analysis of low-carbohydrate and low-fat diets on markers of the metabolic syndrome. Nutr Rev 79: 429–444, 2020. doi: 10.1093/nutrit/nuaa044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Serpico D, Borghini A. From obesity to energy metabolism: ontological perspectives on the metrics of human bodies. Topoi 40: 577–586, 2021. doi: 10.1007/s11245-020-09722-1. [DOI] [Google Scholar]
  • 28. Johnson RK, Appel LJ, Brands M, Howard BV, Lefevre M, Lustig RH, Sacks F, Steffen LM, Wylie-Rosett J. and on behalf of the American Heart Association Nutrition Committee of the Council on Nutrition, Physical Activity, and Metabolism and the Council on Epidemiology and Prevention. Dietary sugars intake and cardiovascular health. Circulation 120: 1011–1020, 2009. doi: 10.1161/CIRCULATIONAHA.109.192627. [DOI] [PubMed] [Google Scholar]
  • 29. Faruque S, Tong J, Lacmanovic V, Agbonghae C, Minaya DM, Czaja K. The dose makes the poison: sugar and obesity in the United States–a review. Pol J Food Nutr Sci 69: 219–233, 2019. doi: 10.31883/pjfns/110735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Magriplis E, Michas G, Petridi E, Chrousos GP, Roma E, Benetou V, Cholopoulos N, Micha R, Panagiotakos D, Zampelas A. Dietary sugar intake and its association with obesity in children and adolescents. Children 8: 676, 2021. doi: 10.3390/children8080676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Bentley RA, Ruck DJ, Fouts HN. U.S. obesity as delayed effect of excess sugar. Econ Hum Biol 36: 100818, 2020. doi: 10.1016/j.ehb.2019.100818. [DOI] [PubMed] [Google Scholar]
  • 32. Skop-Lewandowska A, Zając J, Kolarzyk E. Overweight and obesity vs. simple carbohydrates consumption by elderly people suffering from diseases of the cardiovascular system. Ann Agric Environ Med 24: 575–580, 2017. doi: 10.5604/12321966.1233555. [DOI] [PubMed] [Google Scholar]
  • 33. Stinson EJ, Piaggi P, Ibrahim M, Venti C, Krakoff J, Votruba SB. High fat and sugar consumption during ad libitum intake predicts weight gain. Obesity (Silver Spring) 26: 689–695, 2018. doi: 10.1002/oby.22124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Kim HN, Song SW. Associations between macronutrient intakes and obesity/metabolic risk phenotypes: findings of the Korean National Health and Nutrition Examination Survey. Nutrients 11: 628, 2019. doi: 10.3390/nu11030628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Comuzzie AG, Cole SA, Laston SL, Voruganti VS, Haack K, Gibbs RA, Butte NF. Novel genetics loci identified for the pathophysiology of childhood obesity in the Hispanic population. PLoS One. 7: e51954, 2012. doi: 10.1371/journal.pone.0051954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Pigeyre M, Yazdi FT, Kaur Y, Meyre D. Recent progress in genetics, epigenetics and metagenomics unveils the pathophysiology of human obesity. Clin Sci (Lond) 130: 943–986, 2016. doi: 10.1042/CS20160136. [DOI] [PubMed] [Google Scholar]
  • 37. Kunej T, Jevsinek Skok D, Zorc M, Ogrinc A, Michal JJ, Kovac M, Jiang Z. Obesity gene atlas in mammals. J Genomics 1: 45–55, 2013. doi: 10.7150/jgen.3996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Jiang L, Penney KL, Giovannucci E, Kraft P, Wilson KM. A genome-wide association study of energy intake and expenditure. PLoS One 13: e0201555, 2018. doi: 10.1371/journal.pone.0201555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, , et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 518: 197–206, 2015. doi: 10.1038/nature14177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Celis-Morales CA, Lyall DM, Gray SR, Steell L, Anderson J, Iliodromiti S, Welsh P, Guo Y, Petermann F, Mackay DF, Bailey ME, Pell JP, Gill JM, Sattar N. Dietary fat and total energy intake modifies the association of genetic profile risk score on obesity: evidence from 48170 UK Biobank participants. Int J Obes 41: 1761–1768, 2017. doi: 10.1038/ijo.2017.169. [DOI] [PubMed] [Google Scholar]
  • 41. Choi SW, Mak TS, O’Reilly PF. Tutorial: a guide to performing polygenic risk score analyses. Nat Protoc 15: 2759–2772, 2020. doi: 10.1038/s41596-020-0353-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Lewis CM, Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Med 12: 44, 2020. doi: 10.1186/s13073-020-00742-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Khera AV, Chaffin M, Wade KH, Zahid S, Brancale J, Xia R, Distefano M, Senol-Cosar O, Haas ME, Bick A, Aragam KG, Lander ES, Smith GD, Mason-Suares H, Fornage M, Lebo M, Timpson NJ, Kaplan LM, Kathiresan S. Polygenic prediction of weight and obesity trajectories from birth to adulthood. Cell 177: 587–596.e9, 2019. doi: 10.1016/j.cell.2019.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Singh RK, Kumar P, Mahalingam K. Molecular genetics of human obesity. A comprehensive review. C R Biol 340: 87–108, 2017. doi: 10.1016/j.crvi.2016.11.007. [DOI] [PubMed] [Google Scholar]
  • 45. Goodarzi MO. Genetics of obesity: what genetic association studies have taught us about the biology of obesity and its complications. Lancet Diabetes Endocrinol 6: 223–236, 2018. doi: 10.1016/S2213-8587(17)30200-0. [DOI] [PubMed] [Google Scholar]
  • 46. Young KL, Graff M, Fernandez-Rhodes L, North KE. Genetics of obesity in diverse populations. Curr Diab Rep 18: 145, 2018. doi: 10.1007/s11892-018-1107-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Mariman EC, Bouwman FG, Aller EE, van Baak MA, Wang P. Extreme obesity is associated with variation in genes related to the circadian rhythm of food intake and hypothalamic signaling. Physiol Genomics 47: 225–231, 2015. doi: 10.1152/physiolgenomics.00006.2015. [DOI] [PubMed] [Google Scholar]
  • 48. Valeeva FV, Medvedeva MS, Khasanova KB, Valeeva EV, Kiseleva TA, Egorova ES, Pickering C, Ahmetov II. Association of gene polymorphisms with body weight changes in prediabetic patients. Mol Biol Rep 49: 4217–4224, 2022. doi: 10.1007/s11033-022-07254-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Qi Q, Chu AY, Kang JH, Jensen MK, Curhan GC, Pasquale LR, Ridker PM, Hunter DJ, Willett WC, Rimm EB, Chasman DI, Hu FB, Qi L. Sugar-sweetened beverages and genetic risk of obesity. N Engl J Med. 367: 1387–1396, 2012. doi: 10.1056/NEJMoa1203039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Softic S, Gupta MK, Wang GX, Fujisaka S, O’Neill BT, Rao TN, Willoughby J, Harbison C, Fitzgerald K, Ilkayeva O, Newgard CB, Cohen DE, Kahn CR. Divergent effects of glucose and fructose on hepatic lipogenesis and insulin signaling. J Clin Invest 127: 4059–4074, 2017. doi: 10.1172/JCI94585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Alizadeh S, Pooyan S, Mirzababaei A, Arghavani H, Hasani H, Mirzaei K. Interaction of MC4R rs17782313 variants and dietary carbohydrate quantity and quality on basal metabolic rate and general and central obesity in overweight/obese women: a cross-sectional study. BMC Endocr Disord 22: 121, 2022. doi: 10.1186/s12902-022-01023-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Alsulami S, Nyakotey DA, Dudek K, Bawah AM, Lovegrove JA, Annan RA, Ellahi B, Vimaleswaran KS. Interaction between metabolic genetic risk score and dietary fatty acid intake on central obesity in a Ghanian population. Nutrients 12: 1906, 2020. doi: 10.3390/nu12071906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Corella D, Arnett DK, Tucker KL, Kabagambe EK, Tsai M, Parnell LD, Lai CQ, Lee C, Warodomwichit D, Hopkins PN, Ordovas JM. A high intake of saturated fatty acids strengthens the association between the fat mass and obesity-associated gene and BMI. J Nutr 141: 2219–2225, 2011. doi: 10.3945/jn.111.143826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Kwon YJ, Park DH, Choi JE, Lee D, Hong KW, Lee JW. Identification of the interactions between specific genetic polymorphisms and nutrient intake associated with general and abdominal obesity in middle-aged adults. Clin Nutr 41: 543–551, 2022. doi: 10.1016/j.clnu.2021.12.040. [DOI] [PubMed] [Google Scholar]
  • 55. Primeaux S, Weir A, Denstel K, Martin CK, Hsia D, Staiano A. Role of taste receptor gene variations (SNPs) on diet quality in adolescent females. FASEB J 36: 2022. doi: 10.1096/fasebj.2022.36.S1.R2184. [DOI] [Google Scholar]
  • 56. Park S, Yang HJ, Kim MJ, Hur HJ, Kim SH, Kim MS. Interactions between polygenic risk scores, dietary pattern, and menarche age with the obesity risk in a large hospital-based cohort. Nutrients 13: 3772, 2021. doi: 10.3390/nu13113772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. de Luis D, Aller R, Izaola R, 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 35: 722–730, 2022. doi: 10.1111/jhn.12975. [DOI] [PubMed] [Google Scholar]
  • 58. Franzago M, Di Nicola M, Fraticelli F, Marchioni M, Stuppia L, Vitacolonna E. Nutrigenetic variants and response to diet/lifestyle intervention in obese subjects: a pilot study. Acta Diabetol 59: 69–81, 2022. doi: 10.1007/s00592-021-01787-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Horne JR, Gilliland JA, O’Connor CP, Seabrook JA, Madill J. Change in weight, BMI, and body composition in a population-based intervention versus genetic-based intervention: the NOW Trial. Obesity 28: 1419–1427, 2020. doi: 10.1002/oby.22880. [DOI] [PubMed] [Google Scholar]
  • 60. Garaulet M, Vera B, Bonnet-Rubio G, Gomez-Abellan P, Lee YC, Ordovas JM. Lunch eating predicts weight-loss effectiveness in carriers of the common allele at PERILIPIN1: the ONTIME (Obesity, Nutrigenetics, Timing, Mediterranean) study. Am J Clin Nutr 104: 1160–1166, 2016. doi: 10.3945/ajcn.116.134528. [DOI] [PubMed] [Google Scholar]
  • 61. Aldubayan MA, Pigsborg K, Gormsen SM, Serra F, Palou M, Galmés S, Palou-March A, Favari C, Wetzels M, Calleja A, Rodríguez Gómez MA, Castellnou MG, Caimari A, Galofré M, Suñol D, Escoté X, Alcaide-Hidalgo JM, M Del Bas J, Gutierrez B, Krarup T, Hjorth MF, Magkos F. A double-blinded, randomized, parallel intervention to evaluate biomarker-based nutrition plans for weight loss: the PREVENTOMICS study. Clin Nutr 41: 1834–1844, 2022. doi: 10.1016/j.clnu.2022.06.032. [DOI] [PubMed] [Google Scholar]
  • 62. Yam P, VerHague M, Albright J, Gertz E, de Villena FP, Bennett BJ. Altered macronutrient composition and genetics influence the complex transcriptional network associated with adiposity in the Collaborative Cross. Genes Nutr 17: 13, 2022. doi: 10.1186/s12263-022-00714-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Madkour MI, Malhab LJ, Abdel-Rahman WM, Abdelrahim DN, Saber-Ayad M, Faris ME. Ramadan diurnal intermittent fasting is associated with attenuated FTO gene expression in subjects with overweight and obesity: a prospective cohort study. Front Nutr 8: 741811, 2021. doi: 10.3389/fnut.2021.741811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Aristizabal MJ, Anreiter I, Halldorsdottir T, Odgers CL, McDade TW, Goldenberg A, Mostafavi S, Kobor MS, Binder EB, Sokolowski MB, O’Donnell KJ. Biological embedding of experience: a primer on epigenetics. Proc Natl Acad Sci U S A 117: 23261–23269, 2020. doi: 10.1073/pnas.1820838116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Gallardo-Escribano C, Buonaiuto V, Ruiz-Moreno MI, Vargas-Candela A, Vilches-Perez A, Benitez-Porres J, Romance-Garcia AR, Ruiz-Moreno A, Gomez-Huelgas R, Bernal-Lopez MR. Epigenetic approach in obesity: DNA methylation in a prepubertal population which underwent a lifestyle modification. Clin Epigenetics 12: 144, 2020. doi: 10.1186/s13148-020-00935-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kaspar D, Hastreiter S, Irmler M, Hrabe D, Angelis M, Beckers J. Nutrition and its role in epigenetic inheritance of obesity and diabetes across generations. Mamm Genome 31: 119–133, 2020. doi: 10.1007/s00335-020-09839-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Landecker H. Food as exposure: Nutritional epigenetics and the new metabolism. Biosocieties 6: 167–194, 2011. doi: 10.1057/biosoc.2011.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Li Y. Epigenetic mechanisms link maternal diets and gut microbiome to obesity in the offspring. Front Genet 9: 342, 2018. doi: 10.3389/fgene.2018.00342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Patti ME. Epigenetic mediators of risk for metabolic disease across generations. FASEB J 36: 2022. [Google Scholar]
  • 70. Sasaki A, Murphy KE, Briollais L, McGowan PO, Matthews SG. DNA methylation profiles in the blood of newborn term infants to mothers with obesity. PLoS One 17: e0267946, 2022. doi: 10.1371/journal.pone.0267946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. Maternal high-fat diet induces DNA methylation changes that contribute to glucose intolerance in offspring. Front Endocrinol (Lausanne) 10: 871, 2019. doi: 10.3389/fendo.2019.00871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Schulz L. The Dutch Hunger Winter and the developmental origins of health and disease. Proc Natl Acad Sci U S A 107: 16757–16758, 2010. doi: 10.1073/pnas.1012911107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Bleker LS, de Rooij SR, Painter RC, Ravelli AC, Roseboom TJ. Cohort profile: the Dutch famine birth cohort (DFBC)-a prospective birth cohort study in the Netherlands. BMJ Open 11: e042078, 2021. doi: 10.1136/bmjopen-2020-042078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Veenendaal MV, Painter RC, de Rooij SR, Bossuyt PM, van der Post JA, Gluckman PD, Hanson MA, Roseboom TJ. Transgenerational effects of prenatal exposure to the 1944-45 Dutch famine. BJOG 120: 548–553, 2013. doi: 10.1111/1471-0528.12136. [DOI] [PubMed] [Google Scholar]
  • 75. Meng R, Lv J, Yu C, Bian Z, Yang L, Chen Y, Zhang H, Chen X, Chen J, Chen Z, Chen J, Chen Z, Qi L, lLi L, China Kadoorie Biobank Collaborative Group. Prenatal famine exposure, adulthood obesity patterns and risk of type 2 diabetes. Int J Epidemiol 47: 399–408, 2018. doi: 10.1093/ije/dyx228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Crujeiras AB, Izquierdo AG, Primo D, Milagro FI, Sajoux I, Jacome A, Fernandez-Quintela A, Portillo MP, Martinez JA, Martinez-Olmos MA, de Luis D, Casanueva FF. Epigenetic landscape in blood leucocytes following ketosis and weight loss induced by a very low calorie ketogenic diet (VLCKD) in patients with obesity. Clin Nutr 40: 3959–3972, 2021. doi: 10.1016/j.clnu.2021.05.010. [DOI] [PubMed] [Google Scholar]
  • 77. Bordoni L, Petracci I, Mlodzik-Czyzewska M, Malinowska AM, Szwengiel A, Sadowski M, Gabbianelli R, Chmurzynska A. Mitochondrial DNA and Epigenetics: Investigating interactions with the one-carbon metabolism in obesity. Oxid Med Cell Longev 2022: 9171684, 2022. doi: 10.1155/2022/9171684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Izquierdo AG, Carreira MC, Amil M, Mosteiro CS, Garcia-Caballero T, Fernandez-Quintela A, Portillo MP, Casanueva FF, Crujeiras AB. An energy restriction-based weight loss intervention is able to reverse the effects of obesity on the expression of liver tumor-promoting genes. FASEB J 34: 2312–2325, 2020. doi: 10.1096/fj.201901147RR. [DOI] [PubMed] [Google Scholar]
  • 79. O’Brien J, Hayder H, Zayed Y, Peng C. Overview of microRNA biogenesis, mechanisms of actiones, and circulation. Front Endocrinol 9: 402, 2018. doi: 10.3389/fendo.2018.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Landrier JF, Derghal A, Mounien L. MicroRNAs in obesity and related metabolic disorders. Cells 8: 859, 2019. doi: 10.3390/cells8080859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Formichi C, Nigi L, Grieco GE, Maccora C, Fignani D, Brusco N, Licata G, Sebastiani G, Dotta F. Non-coding RNAs: novel players in insulin resistance and related diseases. Int J Mol Sci 22: 7716, 2021. doi: 10.3390/ijms22147716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Lauria F, Iacomino G, Russo P, Venezia A, Marena P, Ahrens W, De Henauw S, Eiben G, Foraita R, Hebestreit A, Kourides Y, Molnar D, Moreno LA, Veidebaum T, Siani A, I.Family Consortium . Circulating miRNAs are associated with inflammation biomarkers in children with overweight and obesity: results of the I.Family Study. Genes (Basel) 13: 632, 2022. doi: 10.3390/genes13040632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Cantaro G, Filardi T, Sabato C, Vacca A, Migliaccio S, Morano S, Ferretti E. Tissue and circulating microRNAs as biomarkers of response to obesity treatment strategies. J Endocrinol Invest 44: 1159–1174, 2021. doi: 10.1007/s40618-020-01453-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Davis CD. The gut microbiome and its role in obesity. Nutr Today 51: 167–174, 2016. doi: 10.1097/NT.0000000000000167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Aoun A, Darwish F, Hamod N. The influence of the gut microbiome on obesity in adults and the role of probiotics, prebiotics, and synbiotics for weight loss. Prev Nutr Food Sci 25: 113–123, 2020. doi: 10.3746/pnf.2020.25.2.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Dibaise JK, Zhang H, Crowell MD, Krajmalnik-Brown R, Decker A, Rittmann BE. Gut microbiota and its possible relationship with obesity. Mayo Clin Proc 83: 460–469, 2008. doi: 10.4065/83.4.460. [DOI] [PubMed] [Google Scholar]
  • 87. Newman TM, Shively CA, Register TC, Appt SE, Yadav H, Colwell RR, Fanelli B, Dadlani M, Graubics K, Nguyen UT, Ramamoorthy S, Uberseder B, Clear KY, Wilson AS, Reeves KD, Chappell MC, Tooze JA, Cook KL. Diet, obesity, and the gut microbiome as determinants modulating metabolic outcomes in a non-human primate model. Microbiome 9: 100, 2021. doi: 10.1186/s40168-021-01069-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Nie X, Chen J, Ma X, Ni Y, Shen Y, Yu H, Panagiotou G, Bao Y. A metagenome-wide association study of gut microbiome and visceral fat accumulation. Comput Struct Biotechnol J 18: 2596–2609, 2020. doi: 10.1016/j.csbj.2020.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Rampelli S, Guenther K, Turroni S, Wolters M, Veidebaum T, Kourides Y, Molnár D, Lissner L, Benitez-Paez A, Sanz Y, Fraterman A, Michels N, Brigidi P, Candela M, Ahrens W. Pre-obese children’s dysbiotic gut microbiome and unhealthy diets may predict the development of obesity. Commun Biol 1: 222, 2018. doi: 10.1038/s42003-018-0221-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Yan H, Qin Q, Chen J, Yan S, Li T, Gao X, Yang Y, Li A, Ding S. Gut microbiome alterations in patients with visceral obesity based on quantitative computed tomography. Front Cell Infect Microbiol 11: 823262, 2022. doi: 10.3389/fcimb.2021.823262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa S, Navarrete P, Balamurugan R. The Firmicutes/Bacteriodes ratio: a relevant marker of gut dysbiosis in obese patients? Nutrient 12: 1474, 2020. doi: 10.3390/nu12051474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Depommier C, Van Hul M, Everard A, Delzenne NM, de Vos WM, Cani PD. Pasteurized Akkermansia muciniphila increases whole-body energy expenditure and fecal energy excretion in diet-induced obese mice. Gut Microbes 11: 1231–1245, 2020. doi: 10.1080/19490976.2020.1737307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, de Barsy M, Loumaye A, Hermans MP, Thissen JP, de Vos WM, Cani PD. Supplementation with Akkermansia muciniphila in overweight and obese humans volunteers: a proof-of-concept exploratory study. Nat Med 25: 1096–1103, 2019. doi: 10.1038/s41591-019-0495-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Palmas V, Pisanu S, Madau V, Casula E, Deledda A, Cusano R, Uva P, Vascellari S, Loviselli A, Manzin A, Velluzzi F. Gut microbiota markers associated with obesity and overweight in Italian adults. Sci Rep 11: 5532, 2021. doi: 10.1038/s41598-021-84928-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Peters BA, Shapiro JA, Church TR, Miller G, Trinh-Shevrin C, Yuen E, Friedlander C, Hayes RB, Ahn JA. Taxonomic signature of obesity in a large study of American adults. Sci Rep 8: 9749, 2018. doi: 10.1038/s41598-018-28126-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Koh A, Backhed F. From association to causality: the role of the gut microbiota and its functional products on host metabolism. Mol Cell 78: 584–596, 2020. doi: 10.1016/j.molcel.2020.03.005. [DOI] [PubMed] [Google Scholar]
  • 97. Sharma M, Li Y, Stoll ML, Tollefsbol TO. The epigenetic connection between the gut microbiome in obesity and diabetes. Front Genet 10: 1329, 2019. doi: 10.3389/fgene.2019.01329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Woo V, Alenghat T. Epigenetic regulation by gut microbiota. Gut Microbiobes 14: 2022407, 2022. doi: 10.1080/19490976.2021.2022407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Connell J, Ho C, Shen N, Moura P, Cai Y, Tanton D, Banavar G, Vuyisich M. Evidence-based precision nutrition improves clinical outcomes by analyzing human and microbial molecular data with artificial intelligence (Preprint). bioRxiv 2021.04.24.441290, 2021. doi: 10.1101/2021.04.24.441290. [DOI]
  • 100. Lee YC, Christensen JJ, Parnell LD, Smith CE, Shao J, McKeown NM, Ordovas JM, Lai CQ. Using machine learning to predict obesity based on genome-wide and epigenome-wide, gene-gene and gene-diet interactions. Front Genet 12: 783845, 2021. doi: 10.3389/fgene.2021.783845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.National Institutes of Health. NIH awards $170 million for precision nutrition study (Online) Washington, DC: NIH, 2022. https://www.nih.gov/news-events/news-releases/nih-awards-170-million-precision-nutrition-study#:∼:text=The%20National%20Institutes%20of%20Health. [Google Scholar]
  • 102.Centers for Disease Control and Prevention. Prevalence of obesity and severe obesity among adults: United States, 2017–2018 (Online). NCHS Data Brief No. 360, 2020. https://www.cdc.gov/nchs/products/databriefs/db360.htm#:∼:text=Examination%20Survey%20. [Google Scholar]
  • 103. Choi WJ, Shin D. Interactions between red and processed meat consumption and APOA5 gene variants associated with the incidence of metabolic syndrome in Korean adults. Genes Nutr 17: 5, 2022. doi: 10.1186/s12263-022-00707-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Rask-Andersen M, Karlsson T, Ek WE, Johansson Å. Gene-environment interaction study for BMI reveals interactions between genetic factors and physical activity, alcohol consumption and socioeconomic status. PLoS Genet 13: e10069770, 2017. doi: 10.1371/journal.pgen.1006977. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiology are provided here courtesy of American Physiological Society

RESOURCES