You are on page 1of 9

REVIEW PHYSIOLOGY 38: 42–50, 2023. First published September 20, 2022; doi:10.1152/physiol.00014.

2022

Precision Nutrition: Recent Advances in V. Saroja Voruganti


Department of Nutrition and Nutrition Research Institute, University
Obesity of North Carolina at Chapel Hill, Kannapolis, North Carolina
saroja@unc.edu

“Precision nutrition” is an emerging area of nutrition research that focuses on


understanding metabolic variability within and between individuals and helps de-
velop 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 preci-
sion nutrition-based approaches. Recent studies have shown significant associa-
tions 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 environ-
ment (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 metab-
olites, folate, B-vitamins, and short-chain fatty acids interact with miRNAs to influ-
ence 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 obe-
sity and projects the importance of precision nutrition in obesity and weight
management.

epigenetics; metabolic individuality; metabolomics; microbiome; nutrigenomics

Introduction that their energy and dietary needs are different from
each other (3, 4). Using one’s DNA (genetics) to pre-
People have different responses to and requirements dict their response to nutrient intake is only one of the
for nutrients and other components of the diet, and various facets of precision nutrition. Other aspects
this heterogeneity is an important source of variance that influence the variation in individual response to
in nutrition studies. Understanding this variation and nutrients include epigenetic, microbiome, and envi-
its role in risk for diseases has gained more momen- ronmental factors (1, 4, 5).
tum with the advent of new tools and technologies The US National Institutes of Health (NIH) has recog-
(1–3). “Precision nutrition” is an emerging area of nutri- nized and identified precision nutrition as a key area of
tion research that focuses on understanding meta- research in the pursuit of optimal health (6). As an an-
bolic variability within and between individuals and cillary study to their “All of Us” study, the NIH made a
helps to develop customized dietary plans and inter- substantial investment in precision nutrition research,
ventions to maintain optimum individual health. It in recognition that one-size-does-not-fit all with respect
assumes that each person will respond differently and to dietary intake, metabolism, and disease risk (101).

42 1548-9213/23 Copyright © 2023 Int. Union Physiol. Sci./Am. Physiol. Soc.

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
Precision nutrition can be categorized into personal- variants and/or epigenetic modifications, micro-
ized or individualized nutrition and stratified nutrition, bial species and metabolites, lifestyle, and social
the former being at an individual level whereas the lat- and psychological factors, with serious implica-
ter one focusing on groups of susceptible individuals tions for obesity (FIGURE 1) (15–17). This narrative
(4, 7). Precision nutrition is innovative in that it consid- review highlights and discusses the recent advan-
ers individual differences in people’s genes, environ- ces in the components of precision nutrition in
ment, and lifestyles. This information is key for relation to obesity.
targeting treatments for specific group of individuals.
Advances in precision nutrition have led to, and con- Nutritional Genomics and Obesity
tinue to lead to, new discoveries that can provide the
foundations of interventions or treatments tailored to Nutritional genomics (NGx) is the study of how an indi-
specific individuals (7, 8). vidual’s genetics affect the way nutrients are metabo-
Obesity is a major health concern worldwide (9–11). lized (nutrigenetics) and how nutrients affect the way
It poses a considerable burden health-wise and cost- genes are expressed (nutrigenomics) (8, 21). The main
wise. It is also a precursor to various other metabolic aims of NGx are to identify genetic variants that are
diseases such as type 2 diabetes, cardiovascular dis- associated with diet-related diseases, to underlie the
eases, metabolic syndrome, neurodegenerative dis- variability in responses to diet or nutrients, and to help
eases, and some types of cancers (12, 13). As per CDC develop nutritional strategies that can treat or prevent
2017–2018 statistics, 42.4% and 9.2% of adults in the diseases (3, 8, 21). Among the metabolic diseases,
United States have obesity and severe obesity, obesity is the most heavily studied within the NGx par-
respectively (102). The regulation of weight and me- adigm. It is well known that lifestyle factors such as
tabolism including the physiological processes has diet and exercise play a key role in the development
been studied extensively. While the pathogenesis of and treatment of obesity and related comorbidities.
obesity seems as simple as the difference between However, it is also known that weight loss interven-
ingested and expended calories, scientific evidence tions, whether dietary plans or exercise regimens, do
shows that obesity is a complex disease and involves not elicit the same response from all individuals.
genetic, epigenetic, microbiome, psychosocial, and Additionally, the equation of energy in and energy out
environmental factors, as well as the interactions is not as simple as it seems. The interindividual vari-
between them. Additionally, a person’s social, cultural, ability in response to diet or exercise is quite substan-
and behavioral factors also impact a person’s weight tial, even after correcting for differences in age,
and body composition (13, 14). gender, race/ethnicity, and sociodemographic charac-
A key feature of obesity is the dysregulation of teristics (3, 22, 23).
energy metabolism. The central nervous system plays The imbalance between energy intake and
a vital role in appetite, satiety, and energy regulation, energy expenditure is a key determinant of obe-
with the hypothalamus as the master regulator. The sity. Energy intake occurs through intake of mac-
arcuate nucleus (ARC), a part of the hypothalamus, ronutrients, carbohydrates, proteins, and fats. The
is a vital center for the regulation of metabolism and amount and type of these macronutrients deter-
feeding (15–17). It contains neurons that express two mine adiposity and energy equilibrium. A dynamic
distinct, functionally antagonistic, peptides; orexi- time-series analysis of three decades of US data
genic [appetite stimulating: neuropeptide Y (NPY) and a decade of data from 164 other countries, af-
and agouti-related peptide (AgRP)] and anorexi- ter adjustment for total calories, has shown that
genic [appetite suppressing: pro-opiomelanocortin carbohydrate, and not fat, consumption was asso-
(POMC) and cocaine and amphetamine-regulated ciated with short- and long-term increases in
transcript (CART)] peptides. The neuronal signals weight (24). Meta-analysis of studies of low-carbo-
from the lateral hypothalamus (increase food intake) hydrate and low-fat diets for weight loss showed
and parabrachial nucleus (decrease food intake) also that, irrespective of total calories and weight loss,
contribute to the regulation of appetite (18, 19). The pe- reduction in carbohydrates may prove beneficial for
ripheral signals include hormonal signals such as leptin, improving metabolic syndrome markers, including
insulin, and gut hormones [cholecystokinin (CCK), ghre- obesity (25–27).
lin, peptide YY (PYY), glucagon-like peptide1 (GLP1), It is also known that simple or added sugars and sat-
oxyntomodulin, obestatin, and nesfatin] (19, 20). The urated fatty acids can adversely affect adiposity.
integration of hormonal and nutritional signals from the Simple sugars, such as glucose, fructose, lactose, and
circulation and peripheral and central neuronal (includ- sucrose, are metabolized differently than complex car-
ing astrocyte-neuron and microglia-neuron and gut- bohydrates. As per the American Heart Association,
brain axis) signals happens in the ARC to generate a the recommended total sugar consumption for US
feeding response (15–17). Both components of energy adults is 150 and 100 calories per day for men and
metabolism, energy intake and expenditure, have been women, respectively (28). Although much decreased,
shown to be altered or modified by genetic the consumption of simple sugars and saturated fatty

PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org 43

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
acid remains higher than the recommended intake. position and related phenotypes to be 40–70%
Both the monosaccharides glucose and fructose get (35–38). Genome-wide association and candidate
metabolized and yield great amounts of fat in the liver. gene studies have identified several genetic var-
However, they differ in regional adipose deposition iants that affect various components of obesity. In a
with glucose promoting subcutaneous adipose deposi- study of more than 300,000 European adults,
tion while fructose favors visceral adipose tissue depo- Locke et al. (39) identified 97 loci associated with
sition (29). Other studies have shown the adverse body mass index (BMI). Many of these loci, particu-
effects of simple sugars, either fructose or glucose, on larly a specific set of 32 loci, have been consistently
adiposity and weight gain (30–33). However, in a reported to be associated with obesity across eth-
recent study investigating the association between nicities. Some studies have generated polygenic risk
macronutrient intakes and obesity and metabolic risk, scores (PRS) from these 32 loci and found stronger
the authors did not find any link between carbohy- effects on anthropometric measures (40). The PRS is
drates, fats, and weight (34). These studies and results computed from an independent set of polymorphisms
show differences in the way nutrients affect obesity that are associated with the risk for a disease. The
phenotypes and point to the involvement of other bio- total number of risk alleles per individual is summed,
logical and environmental factors. weighted or unweighted by its effect size, and a score
Individual variability in weight phenotypes has a is calculated. A single score, the PRS, thus reflects the
strong genetic basis. Twin, family-based, and other cumulative effect of the included polymorphisms and
studies have estimated heritabilities of body com- is a better predictor of the disease risk (41, 42). A study

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.

44 PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
in the UK Biobank has generated a PRS with 2.1 million program with genetic testing. The latter group showed
single nucleotide polymorphisms (SNPs) associated better weight loss than the former one for as long as 6
with obesity and assessed them in individuals ranging months (59). Similar results were shown in another trial
from birth to middle age (43). that had 284 individuals who underwent a high-fat,
Prominent among the genes that are associated hypocaloric diet. They found greater weight loss in T
with appetite or adiposity are fat mass and obesity carriers of rs10401670 of RETN as compared to others
related (FTO), melanocortin 4 receptor 4 (MC4R), apoli- (57). Studies have also shown the effect of a few
poprotein A5 (APOA5), neuropeptide Y (NPY), leptin genetic variants on the extent of weight loss in inter-
(LEP), leptin receptor (LEPR), brain-derived neurotro- vention studies. A study by Franzago et al. (58) found
phic factor (BDNF), single-minded homolog1 (SIM1), that A-allele carriers of rs9939609 of the FTO gene
neurexin3 (NRXN3), NPC intracellular cholesterol lost less weight and decreased BMI less than TT car-
transporter 1 (NPC1), neuronal growth regulator q riers during a 12-mo diet/lifestyle intervention. A study
(NEGR1), mitochondrial carrier homolog 2 (MTCH2), by Garaulet et al. (60) found that eating late was linked
apolipoprotein E (APOE), tumor necrosis factor alpha to lower weight loss in AA genotype of PLIN1
(TNFa), plasminogen activator inhibitor 1 (PAI-1), trans- 14995 A > T carriers, thus pointing to the importance
membrane protein 18 (TMEM18), etc. (43–46). A study of meal timing along with genes in obesity. On the
investigating genetic influence on extreme obesity other hand, a 10-wk weight-loss intervention based on
found several loci that are linked to either the circa- 35 SNPs did not find any difference between person-
dian rhythm of food consumption or hypothalamic sig- alized dietary intervention based on 35 SNPs and a
naling related to food intake (47, 48). generic healthy diet with respect to either loss of body
Studies have also shown differential effects of weight or fat mass (61).
nutrients based on an individual’s genetic makeup. In a similar vein, modified nutrient diets have been
Genetic variants interact with nutrition and other envi- linked to changes in the expression of genes associ-
ronmental factors such as physical activity to affect ated with adiposity. In a collaborative cross mice
obesity phenotypes. The most frequently studied model, Yam et al (62) showed that genetic back-
nutrients have been simple sugars and saturated and ground and macronutrient alterations affected the
unsaturated fatty acids. Simple sugars such as fruc- expression of genes involved in adiposity. There is an
tose and glucose, either in the form of sugar-sweet- increasing interest in intermittent fasting and its effect
ened beverages (SSB) or as part of high fructose/ on metabolic diseases including obesity. A study of
glucose diets, have been studied to understand their individuals with overweight/obesity showed a signifi-
effects on obesity and related comorbidities (49, 50). cant decrease in FTO gene expression after the
In circulation, fructose and glucose have different met- month-long fasting period of Ramadan in contrast to
abolic fates, although fructose needs glucose for its normal weight individuals (63).
metabolic effects. Fructose, more than glucose, favors
hepatic lipogenesis (49). A large study including 6,934 Nutritional Epigenetics and Obesity
women from the Nurses Health Study, 4,423 men
from the Health Professionals Follow-up Study, and Epigenetics refer to the genetic effects that are exter-
21,740 women from the Women’s Genome Health nal to changes in DNA sequence. The various types of
Study generated a PRS, based on the aforementioned epigenetic changes include DNA methylation (addi-
32 obesity-associated SNPs. They found that with tion of methyl groups to cysteine or adenine on the
an increment per 10 risk alleles, the relative risk for DNA), noncoding RNAs (short and long noncoding
BMI increased with every category of increasing SSB RNAs that modulate mRNAs to affect gene expres-
intake (49). Some other examples of gene by diet sion), and histone modification (posttranslational modi-
interactions with respect to obesity include carbohy- fication of histone proteins that can affect chromatin
drate intake and MC4R rs17782313 (51), low-protein function and DNA accessibility) (64–66). A branch of
diet and genetic risk score (GRS) of 15 SNPs (52), satu- epigenetics, nutritional epigenetics, is the study of the
rated fat intake and FTO (53), vitamin C intake and car- effects of food on the expression of genes (67).
boxy peptidase (CP) rs59465035 (54), fat and sugar Human and animal studies have shown that epige-
intake and taste receptor (TAS1R2) (55), 5 SNP PRS netic effects can transcend generations. Maternal
and fried foods (56), high-fat hypocaloric diet and nutrition, in particular, can have distinct epigenetic
resistin (RETN) rs10401670 (57), etc. (Table 1). effects on the child’s risk for various metabolic dis-
Weight loss interventions have also shown better eases (68–70).
results when participant genetics were considered. An individual’s alleles are determined by their
For example, a randomized controlled clinical trial, maternal or paternal origins (genomic imprinting),
The Nutrigenomics, Overweight/Obesity and Weight and these are involved in a variety of metabolic
Management (NOW) study conducted a weight loss functions. Alterations in genomic imprinting due to
trial with two groups, one with a lifestyle balance pro- epigenetic mechanisms such as DNA methylation,
gram and the other one with a lifestyle balance histone modifications, or genetic events such as

PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org 45

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
TABLE 1. Summary of nutrigenetics of obesity phenotypes
Variant of Nutrient/Dietary
Interest Gene Type of Study Sample Size Intake Outcome Population Conclusion Reference(s)

rs9939609 FTO Cohort 339,000 Total fat BMI European and TT allele of both SNPs asso- Corella et al. (53)
rs1121980 Hispanic adults ciated with higher BMI
with higher SFA intake
rs9939609 FTO Intervention 86 Mediterranean Weight loss Italian A allele was associated with Franzago et al.
diet less weight loss than TT (58)
carriers
rs662799 APOA5 Cohort 3266 Red meat Metabolic Korean G carriers and in highest ter- Choi and Shin,
syndrome tile of red meat consump- (103)
tion had 1.7 HR as
compared to those with A
allele
GRS of 4 SNPs Cohort 302 Total fat, SFA, Waist Ghana GRS >3 and high intake of Alsulami et al.
MUFA, and circumference fat, SFA, PUFA, and (52)
PUFA MUFA linked to increased
waist circumference
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 Increase in BMI per GRS is Rask-Anderson et
Biobank) higher in infrequent al. (104)
drinkers as compared to
frequent drinkers
rs6722579 CAB39 Cohort 50,808 Fat intake Abdominal Korea A carriers were at higher Kwon et al. (54)
obesity risk for abdominal obesity
in those who had fat
intake >DRI
rs59465035 CPQT T carriers are at lower risk
for abdominal obesity in
those who had vitamin C
>DRI
rs17782313 MC4R Cohort 282 Carbohydrate BMI Iranian Higher carbohydrate intake Alizadeh et al. (51)
intake was associated with
higher BMI and waist cir-
cumference in C carriers
rs1801282 PPARG Intervention 327 Diet therapy vs. Weight loss CC carriers had a greater Valeeva et al. (48)
diet therapy þ weight loss than others
metformin
rs59465035 CP Cohort 50,808 Vitamin C Waist Korean T-allele carriers had lower Kwon et al. (54)
circumference risk of abdominal obesity
as compared to others in
those who consumed
vitamin C higher than DRI
rs10401670 RETN Intervention 284 High-fat hypo- BMI Spain T-allele carriers had greater De Luis et al. (57)
caloric diet weight loss than others
5 SNP PRS Cohort >53,000 Fried foods BMI Korean High PRS and menarche Park et al. (56)
linked with higher BMI
rs1761667, CD36 Cohort 142 Fat and sugar BMI percentiles African American AA and altered fat percep- Primeaux et al.
intake females tion linked to BMI (55)
increase
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.

translocation, inversion, duplication, etc., can result results were shown in studies using data from the
in obesity (67). Animal studies have shown that pa- Chinese Famine that occurred between 1959 and
ternal BMI at the time of conception can influence 1961. Individuals born during this period reported
an infant’s birth weight and patterns of DNA methyl- higher cases of obesity, type 2 diabetes, and meta-
ation in infant cord blood (71). Several studies have bolic syndrome as compared to those born after the
evaluated the Dutch Famine Birth Cohort of the famine (74, 75). In another human study, DNA col-
Hunger Winter. These studies showed that intrauter- lected from infants born at term with 40 women of
ine exposure to maternal undernutrition results in normal BMI and with obesity revealed genomic loci
long-term effects on a child’s metabolic health, associated with differentially methylated regions
including obesity, type 2 diabetes, and metabolic (DMRs) in CpG-dense regions (70). When compared
syndrome (72, 73). Interestingly, these exposures with another larger study, there was a significant
had long-term transgenerational effects. Similar overlap in DMRs, with key genes being PTPRN2, a

46 PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
gene involved in insulin secretion, and MAD1L1, a gene energy from fermented residual peptides and pro-
involved in cell cycle and tumor progression (70). teins. The microorganisms residing in the gut have
An intervention study that included 12 months of life- been associated with obesity risk (85, 86). Individuals
style modification (Mediterranean diet and physical with obesity have specific alterations in the compo-
activity) showed links between BMI and DNA methyla- sition and function of the gut microbiome (86–88),
tion patterns. DNA methylation patterns in genes and the gut microbiome affects energy balance by
related to lipid metabolism and inflammation were the affecting both energy utilization from the diet as
most modified by lifestyle interventions (65). In yet well as energy expenditures and storage. The
another intervention study that involved a very-low- effects of gut microbiome on obesity phenotype
calorie diet (VLCD), the investigators found that after a and weight loss have been captured through popu-
VLCD therapy in individuals with obesity, DNA methyl- lation-based cohort studies as well as randomized
ation patterns changed and resembled more those of controlled trials (89, 90).
a normal weight person (76). In a cohort of normal- Studies have shown that individuals with obesity
weight and overweight-obese individuals, higher BMI have a greater amount of Firmicutes and higher
was associated with lower methylenetetrahydrofolate Firmicutes to Bacteriodes ratios (91). Other studies
reductase (MTHFR) gene methylation (77). Obesity- have shown Firmicutes and Bacteriodes to be inver-
associated alterations in methylation were also sely and positively associated with energy expendi-
observed in individuals on either weight-loss diets or ture and percent body fat, respectively (63). A recent
surgeries. In a group of individuals undergoing either study by Depommier et al. (92) showed that pasteur-
a very-low-calorie ketogenic diet (VLCKD), a balanced ized Akkermansia muciniphila increased energy ex-
hypocaloric diet (BHD), or bariatric surgery, DNA meth- penditure and reduced body weight and fat mass in
ylation in the angiotensin-converting enzyme (ACE2) diet-induced obesity in mice models. The same
was higher in individuals with obesity as compared to groups showed that Akkermansia also improved gut
normal-weight individuals. VLCKD and BHDs helped barrier integrity, insulin resistance, and dyslipidemia in
reverse this hypermethylation, but bariatric surgery mice. They also translated these findings to humans in
did not show a similar benefit (78). an exploratory study where they found a reduction
Noncoding RNA, especially microRNAs (miRNA), is in markers of liver dysfunction and inflammation after
another class of epigenetic markers that have been supplementing the diet with Akkermansia (93). Similar
linked to obesity and risk for other metabolic diseases results have been shown in humans using other forms
(79). These miRNA can affect mRNA stability and deg- of bacteria (88, 90, 93). In a study in normal weight
radation by binding anywhere along the length of the individuals with high and low visceral fat, the inves-
mRNA transcript (80, 81). Like genetic variants, several tigators found that a total of 16 species of microbes
loci that correspond to miRNAs have been identified. were significantly correlated with visceral fat accu-
Kunej et al. (37) identified 1,736 genomic loci associ- mulation as measured by quantitative computed to-
ated with obesity, of which 221 correspond to miRNAs. mography but not with BMI or waist circumference
Several studies have also found these miRNA to be (90). Of these Bacteriodes were associated with
correlated with diet and lifestyle (82, 83), key ones low visceral fat. A longitudinal study of obesity con-
being miR-17/20/93, 21-590-5p, 200 b/c, 221/222, let- ducted in individuals undergoing laparoscopic
7/miR-98, and miR-203 families of miRNAs. In a chil- sleeve gastrectomy (LSG), found a negative correla-
dren study, sex-specific association with obesity was tion between visceral fat and few microbial species,
found with miRNAs, 26 b-3p, hsa-576-5p, hsa-31-5p, the strongest being with Eubacterium eligens.
hsa-10b-5p, and hsa-31-5p. (82). The understanding of Before and after LSG analysis showed a significant
the human epigenome and its interaction with envi- increase in specific species such as C. symbiosum,
ronment, particularly diet, may provide greater insight C. hathewayi, C. citroniae, and other Clostridiales
into the pathogenesis of obesity and help develop after LSG (88). This is significant as Clostridiales are
personalized nutritional interventions. considered to be “good” microbes and are nega-
tively associated with metabolic diseases (94).
Microbiome and Obesity The gut microbiota, whose genome is more than
100 times the human genome, are also known to pass
The human body contains millions of microbes that from the mother to the neonate through exposure dur-
can have significant health effects. The microorgan- ing delivery and through the breastfeeding (95). In
isms, collectively termed the microbiome, are from addition, the microbial metabolites such as short-chain
bacterial, fungal, and protozoal origins (84). The com- fatty acids, trimethylamine-N-oxide (TMAO), folate,
position of gut microbiota is influenced, to a greater and other B vitamins, etc., are known to modulate epi-
extent, by indigestible foods. The microbiota residing genetic mechanisms including DNA methylation, his-
in GI tracts prefer fermentable carbohydrates as sub- tone modifications, and noncoding RNAs (96, 97). The
strates whereas they use proteins and amino acids for microbiota is increasingly being recognized as a
their own enzymes. The distant colon thus gains potential synthesizer of biological compounds that

PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org 47

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
can be used as epigenetic substrates or regulators of individualized or stratified, is bright and can be an
epigenetic enzyme activity (98). effective adjunct therapy to existing treatment strat-
egies. n
Conclusions
V. S. Voruganti is funded by the National Institute of
Precision nutrition is critical to formulating effective di- Diabetes and Digestive and Kidney Diseases Grants
R01DK126666 and P30DK056350, and Eunice Kennedy
etary plans customized for individuals or a group of Shriver National Institute of Child Health and Human
susceptible individuals of diverse ethnicities and pop- Development Grant UG1HD107692.
ulations. To develop personalized or stratified dietary No conflicts of interest, financial or otherwise, are
plans, either to maintain optimal health or to treat met- declared by the authors.
abolic diseases, there is a need to understand how an V.S.V. conceived and designed research; V.S.V. drafted
manuscript; V.S.V. edited and revised manuscript; V.S.V.
individual responds to diet or nutrients and the factors approved final version of manuscript.
responsible for this variation in response. These areas
of nutrition fall under the purview of precision nutrition. References
Knowledge of genetic variants affecting nutrient meta-
1. Toro-Martín J, Arsenault B, Despr es JP, Vohl MC. Precision
bolic pathways is not new but dates to the recognition Nutrition: a review of personalized nutritional approaches for the
of phenylketonuria, lactase persistence, glucose phos- prevention and management of Metabolic Syndrome. Nutrient 9:
913, 2017. doi:10.3390/nu9080913.
phate dehydrogenase deficiency, etc. What is new is
2. Zeisel SH. A conceptual framework for studying and investing in
that with the advent of next-generation sequencing precision nutrition. Front Genet 10: 200, 2019. doi:10.3389/
technologies and the latest analytical approaches, we fgene.2019.00200.

are now able to conduct a comprehensive investiga- 3. Berry SE, Valdes AM, Drew DA, Asnicar F, Mazidi M, Wolf J,
Capdevila J, Hadjigeorgiou G, Davies R, Al Khatib H, Bonnett C,
tion using ‘Omics’ approaches, which are revolutioniz- Ganesh S, Bakker E, Hart D, Mangino M, Merino J, Linenberg I,
ing the field. However, there are some issues that Wyatt P, Ordovas JM, Gardner CD, Delahanty LM, Chan AT,
Segata N, Franks PW, Spector TD. Human postprandial
need to be resolved before precision nutrition can be responses to food and potential for precision nutrition. Nat
Med 26: 964--973, 2020. doi:10.1038/s41591-020-0934-0.
put into practice. First, self-reporting approaches con-
tinue to be the primary methods for collecting dietary 4. Zeisel SH. Precision (Personalized) nutrition: understanding met-
abolic heterogeneity. Annu Rev Food Sci Technol 11: 71--92,
data. Unless there is an objective approach to meas- 2020. doi:10.1146/annurev-food-032519-051736.
uring dietary intake, accurate estimates of dietary 5. Kirk D, Catal C, Tekinerdogan B. Precision Nutrition: a system-
intake (especially long term) will be difficult. Second, atic literature review. Comput Biol Med 133: 104365, 2021.
doi:10.1016/j.compbiomed.2021.104365.
there is a need to better integrate multiple sources of
6. Rodgers GP, Collins FS. Precision Nutrition--the answer to ’’what
data. Usage of machine learning approaches is help- to eat to stay healthy’’. JAMA 324: 735--736, 2020. doi:10.1001/
ing address the integration of multiple sources of jama.2020.13601.

data. Third, proper flow of data among researchers, 7. Mullins VA, Bresette W, Johnstone L, Hallmark B, Chilton FH.
Genomics in personalized nutrition: can you ’’eat for your gen-
practitioners, and educators is needed for dissemina- es’’? Nutrients 12: 3118, 2020. doi:10.3390/nu12103118.
tion or translation of these results. For example, physi- 8. Marcum JA. Nutrigenetics/nutrigenomics, personalized nutrition
cians and dietitian need proper training to be able to and precision healthcare. Curr Nutr Rep 9: 338--345, 2020.
doi:10.1007/s13668-020-00327-z.
prescribe genotype-guided diet plans to patients.
9. Bluher M. Obesity: global epidemiology and pathogenesis.
Fourth, proper counseling of patients is needed for Nat Rev Endocrinol 15: 288--298, 2019. doi:10.1038/s41574-
them to be able to comply with the personalized diet. 019-0176-8.
Despite all these caveats, incorporation of precision 10. Safaei M, Sundararajan EA, Driss M, Boulila W, Shapi'i A. A sys-
tematic literature review on obesity: undertstanding the causes
nutrition approaches has started showing noteworthy and consequences of obesity and reviewing various machine
results. learning approaches used to predict obesity. Comp Biol Med
136: 104754, 2021. doi:10.1016/j.compbiomed.2021.104754.
In a study conducted by Connell et al. (99), research-
11. Gjermeni E, Kirstein AS, Kolbig F, Kirchhof M, Bundalian L,
ers provided participants with personalized data- Katzmann JL, Laufs U, Bluher M, Garten A, Le Duc D. Obesity-an
driven dietary recommendations, using artificial intelli- update on the basic pathophysiology and review of recent thera-
peutic advances. Biomolecules 11: 1426, 2021. doi:10.3390/
gence, and found significant improvement in clinical biom11101426.
outcomes in all conditions and disease activities. 12. Herrera BM, Keildson S, Lindgren CM. Genetics and epige-
Using dietary, genetic, and epigenetic data and netics of obesity. Maturitas 69: 41--49, 2011. doi:10.1016/j.
maturitas.2011.02.018.
machine learning approaches, Lee et al. (100) identi-
13. Rohde K, Keller M, la Cour Poulsen L, Bluher M, Kovacs P,
fied 21 SNPs, 230 DNA methylation sites in lipid and Bottcher Y. Genetics and epigenetics of obesity. Metabolism 92:
carbohydrate metabolism-related genes, and 26 die- 37--50, 2019. doi:10.1016/j.metabol.2018.10.007.
tary factors that predicted obesity with 70% accuracy. 14. Bouchard C. Genetics of obesity: what have we learned over
decades of research. Obesity (Silver Spring) 29: 802--820, 2021.
The main goal of precision nutrition is to understand doi:10.1002/oby.23116.
the differences in the metabolic responses to diet/ 15. Timper K, Bruning JC. Hypothalamic circuits regulating appetite
nutrient intake between individuals and develop tai- and energy homeostasis: pathways to obesity. Dis Model Mech
10: 679--689, 2017. doi:10.1242/dmm.026609.
lored or customized nutritional plans for optimal
health. With the latest Omics and machine-learning- 16. Seong J, Kang JY, Sun JS, Kim KW. Hypothalamic inflammation
and obesity: a mechanistic review. Arch Pharm Res 42: 383--
based approaches, the future of precision nutrition, 392, 2019. doi:10.1007/s12272-019-01138-9.

48 PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
17. Rodriguez-Rodriguez R, Miralpeix C. 33. Stinson EJ, Piaggi P, Ibrahim M, Venti C, Krakoff J, 49. Qi Q, Chu AY, Kang JH, Jensen MK, Curhan GC,
Hypothalamic regulation of obesity. Int J Mol Sci Votruba SB. High fat and sugar consumption during Pasquale LR, Ridker PM, Hunter DJ, Willett WC,
22: 13459, 2021. doi:10.3390/ijms222413459. ad libitum intake predicts weight gain. Obesity Rimm EB, Chasman DI, Hu FB, Qi L. Sugar-sweet-
(Silver Spring) 26: 689--695, 2018. doi:10.1002/ ened beverages and genetic risk of obesity. N
18. Molfino A, Imbimbo G. Editorial: appetite control in oby.22124. Engl J Med. 367: 1387--1396, 2012. doi:10.1056/
obesity. Front Nutr 9: 959627, 2022. doi:10.3389/ NEJMoa1203039.
fnut.2022.959627. 34. Kim HN, Song SW. Associations between macronu-
trient intakes and obesity/metabolic risk pheno- 50. Softic S, Gupta MK, Wang GX, Fujisaka S, O’Neill
19. Miller GD. Appetite regulation: hormones, peptides types: findings of the Korean National Health and BT, Rao TN, Willoughby J, Harbison C, Fitzgerald K,
and neurotransmitters and their role in obesity. Am Nutrition Examination Survey. Nutrients 11: 628, Ilkayeva O, Newgard CB, Cohen DE, Kahn CR.
J Lifestyle Med 13: 586--601, 2019. doi:10.1177/ 2019. doi:10.3390/nu11030628. Divergent effects of glucose and fructose on he-
1559827617716376. patic lipogenesis and insulin signaling. J Clin Invest
35. Comuzzie AG, Cole SA, Laston SL, Voruganti VS, 127: 4059--4074, 2017. doi:10.1172/JCI94585.
20. Myers MG Jr, Olson DP. Central nervous system Haack K, Gibbs RA, Butte NF. Novel genetics loci
control of metabolism. Nature 491: 357--363, 2012. identified for the pathophysiology of childhood obe- 51. Alizadeh S, Pooyan S, Mirzababaei A, Arghavani
doi:10.1038/nature11705. sity in the Hispanic population. PLoS One. 7: H, Hasani H, Mirzaei K. Interaction of MC4R
e51954, 2012. doi:10.1371/journal.pone.0051954. rs17782313 variants and dietary carbohydrate quan-
21. Doo M, Kim Y. Obesity: interactions of genome tity and quality on basal metabolic rate and general
and nutrients intake. Prev Nutr Food Sci 20: 1--7, 36. Pigeyre M, Yazdi FT, Kaur Y, Meyre D. Recent pro-
and central obesity in overweight/obese women: a
2015. doi:10.3746/pnf.2015.20.1.1. gress in genetics, epigenetics and metagenomics
cross-sectional study. BMC Endocr Disord 22: 121,
unveils the pathophysiology of human obesity.
2022. doi:10.1186/s12902-022-01023-5.
22. Hattis D, Banati P, Goble R, Burmaster DE. Human Clin Sci (Lond) 130: 943--986, 2016. doi:10.1042/
interindividual variability in parameters related to CS20160136. 52. Alsulami S, Nyakotey DA, Dudek K, Bawah AM,
health risks. Risk Anal 19: 711--726, 1999. doi:10.1111/ Lovegrove JA, Annan RA, Ellahi B, Vimaleswaran
37. Kunej T, Jevsinek Skok D, Zorc M, Ogrinc A, Michal
j.1539-6924.1999.tb00441.x. JJ, Kovac M, Jiang Z. Obesity gene atlas in mam- KS. Interaction between metabolic genetic risk
mals. J Genomics 1: 45--55, 2013. doi:10.7150/ score and dietary fatty acid intake on central obesity
23. Barber-Chaomoux N, Milenkovic D, Verny MA, in a Ghanian population. Nutrients 12: 1906, 2020.
Habauzit V, Pereira B, Lambert C, Richard D, Boby jgen.3996.
doi:10.3390/nu12071906.
C, Mazur A, Lusson JR, Dobray C, Morand C. 38. Jiang L, Penney KL, Giovannucci E, Kraft P, Wilson
Substantial variability across individuals in the vas- KM. A genome-wide association study of energy 53. Corella D, Arnett DK, Tucker KL, Kabagambe EK,
cular and nutrigenomic response to an acute intake intake and expenditure. PLoS One 13: e0201555, Tsai M, Parnell LD, Lai CQ, Lee C, Warodomwichit
of curcumin: a randomized controlled trial. Mol Nutr 2018. doi:10.1371/journal.pone.0201555. D, Hopkins PN, Ordovas JM. A high intake of satu-
Food Res 62: 1700418, 2018. doi:10.1002/mnfr. rated fatty acids strengthens the association
201700418. 39. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, between the fat mass and obesity-associated gene
Day FR, et al. Genetic studies of body mass index and BMI. J Nutr 141: 2219--2225, 2011. doi:10.3945/
24. Reira-Crichton D, Teft N. Macronutrients and obe- yield new insights for obesity biology. Nature 518: jn.111.143826.
sity: revisiting the calories in, calories out frame- 197--206, 2015. doi:10.1038/nature14177.
work. Econ Hum Biol 14: 33--49, 2014. doi:10.1016/j. 54. Kwon YJ, Park DH, Choi JE, Lee D, Hong KW, Lee
ehb.2014.04.002. 40. Celis-Morales CA, Lyall DM, Gray SR, Steell L, JW. Identification of the interactions between spe-
Anderson J, Iliodromiti S, Welsh P, Guo Y, cific genetic polymorphisms and nutrient intake
25. San-Cristobal R, Navas-Carretero S, Martinez- Petermann F, Mackay DF, Bailey ME, Pell JP, Gill associated with general and abdominal obesity in
Gonzalez MA, Ordovas JM, Martinez JA. Contribution JM, Sattar N. Dietary fat and total energy intake middle-aged adults. Clin Nutr 41: 543--551, 2022.
of macronutrients to obesity: implications for pre- modifies the association of genetic profile risk score doi:10.1016/j.clnu.2021.12.040.
cision nutrition. Nat Rev Endocrinol 16: 305--320, on obesity: evidence from 48170 UK Biobank partic-
2020. doi:10.1038/s41574-020-0346-8. ipants. Int J Obes 41: 1761--1768, 2017. doi:10.1038/ 55. Primeaux S, Weir A, Denstel K, Martin CK, Hsia D,
ijo.2017.169. Staiano A. Role of taste receptor gene variations
26. Willems AE, Jong MS, van Beek AP, Nederhof E, (SNPs) on diet quality in adolescent females. FASEB
van Dijk G. Effects of macronutrient intake in obe- 41. Choi SW, Mak TS, O’Reilly PF. Tutorial: a guide to J 36: 2022. doi:10.1096/fasebj.2022.36.S1.R2184.
sity: a meta-analysis of low-carbohydrate and low- performing polygenic risk score analyses. Nat
fat diets on markers of the metabolic syndrome. Protoc 15: 2759--2772, 2020. doi:10.1038/s41596- 56. Park S, Yang HJ, Kim MJ, Hur HJ, Kim SH, Kim MS.
020-0353-1. Interactions between polygenic risk scores, dietary
Nutr Rev 79: 429--444, 2020. doi:10.1093/nutrit/
pattern, and menarche age with the obesity risk in a
nuaa044. 42. Lewis CM, Vassos E. Polygenic risk scores: from large hospital-based cohort. Nutrients 13: 3772,
research tools to clinical instruments. Genome Med 2021. doi:10.3390/nu13113772.
27. Serpico D, Borghini A. From obesity to energy me-
12: 44, 2020. doi:10.1186/s13073-020-00742-5.
tabolism: ontological perspectives on the metrics
57. de Luis D, Aller R, Izaola R, Primo D. Role of the
of human bodies. Topoi 40: 577--586, 2021. 43. Khera AV, Chaffin M, Wade KH, Zahid S, Brancale J, rs10401670 variant in the resistin gene on the meta-
doi:10.1007/s11245-020-09722-1. Xia R, Distefano M, Senol-Cosar O, Haas ME, Bick A, bolic response after weight loss secondary to a
Aragam KG, Lander ES, Smith GD, Mason-Suares H, high-fat hypocaloric diet with a Mediterranean pat-
28. Johnson RK, Appel LJ, Brands M, Howard BV, Fornage M, Lebo M, Timpson NJ, Kaplan LM,
Lefevre M, Lustig RH, Sacks F, Steffen LM, Wylie- tern. J Hum Nutr Diet 35: 722--730, 2022. doi:10.1111/
Kathiresan S. Polygenic prediction of weight and jhn.12975.
Rosett J. and on behalf of the American Heart obesity trajectories from birth to adulthood. Cell 177:
Association Nutrition Committee of the Council 587--596.e9, 2019. doi:10.1016/j.cell.2019.03.028. 58. Franzago M, Di Nicola M, Fraticelli F, Marchioni M,
on Nutrition, Physical Activity, and Metabolism Stuppia L, Vitacolonna E. Nutrigenetic variants and
and the Council on Epidemiology and Prevention. 44. Singh RK, Kumar P, Mahalingam K. Molecular genet- response to diet/lifestyle intervention in obese sub-
Dietary sugars intake and cardiovascular health. ics of human obesity. A comprehensive review. C R jects: a pilot study. Acta Diabetol 59: 69--81, 2022.
Circulation 120: 1011--1020, 2009. doi:10.1161/ Biol 340: 87--108, 2017. doi:10.1016/j.crvi.2016.11. doi:10.1007/s00592-021-01787-7.
CIRCULATIONAHA.109.192627. 007.
59. Horne JR, Gilliland JA, O’Connor CP, Seabrook JA,
29. Faruque S, Tong J, Lacmanovic V, Agbonghae C, 45. Goodarzi MO. Genetics of obesity: what genetic Madill J. Change in weight, BMI, and body composi-
Minaya DM, Czaja K. The dose makes the poison: association studies have taught us about the biol-
tion in a population-based intervention versus
sugar and obesity in the United States--a review. Pol ogy of obesity and its complications. Lancet
genetic-based intervention: the NOW Trial. Obesity
J Food Nutr Sci 69: 219--233, 2019. doi:10. Diabetes Endocrinol 6: 223--236, 2018. doi:10.1016/
28: 1419--1427, 2020. doi:10.1002/oby.22880.
31883/pjfns/110735. S2213-8587(17)30200-0.
60. Garaulet M, Vera B, Bonnet-Rubio G, Gomez-
30. Magriplis E, Michas G, Petridi E, Chrousos GP, 46. Young KL, Graff M, Fernandez-Rhodes L, North
Abellan P, Lee YC, Ordovas JM. Lunch eating pre-
KE. Genetics of obesity in diverse populations.
Roma E, Benetou V, Cholopoulos N, Micha R, dicts weight-loss effectiveness in carriers of the
Curr Diab Rep 18: 145, 2018. doi:10.1007/s11892-
Panagiotakos D, Zampelas A. Dietary sugar intake common allele at PERILIPIN1: the ONTIME
018-1107-0.
and its association with obesity in children and (Obesity, Nutrigenetics, Timing, Mediterranean)
adolescents. Children 8: 676, 2021. doi:10.3390/ 47. Mariman EC, Bouwman FG, Aller EE, van Baak study. Am J Clin Nutr 104: 1160--1166, 2016.
children8080676. MA, Wang P. Extreme obesity is associated with doi:10.3945/ajcn.116.134528.
variation in genes related to the circadian rhythm
31. Bentley RA, Ruck DJ, Fouts HN. U.S. obesity as of food intake and hypothalamic signaling. 61. Aldubayan MA, Pigsborg K, Gormsen SM, Serra F,
delayed effect of excess sugar. Econ Hum Biol 36: Physiol Genomics 47: 225--231, 2015. doi:10.1152/ Palou M, Galm es S, Palou-March A, Favari C,
100818, 2020. doi:10.1016/j.ehb.2019.100818. physiolgenomics.00006.2015. Wetzels M, Calleja A, Rodríguez G omez MA,
Castellnou MG, Caimari A, Galofr e M, Su~ nol D,
32. Skop-Lewandowska A, Zajaôc J, Kolarzyk E. 48. Valeeva FV, Medvedeva MS, Khasanova KB, Escot e X, Alcaide-Hidalgo JM, M Del Bas J,
Overweight and obesity vs. simple carbohydrates Valeeva EV, Kiseleva TA, Egorova ES, Pickering C, Gutierrez B, Krarup T, Hjorth MF, Magkos F. A dou-
consumption by elderly people suffering from dis- Ahmetov II. Association of gene polymorphisms ble-blinded, randomized, parallel intervention to
eases of the cardiovascular system. Ann Agric with body weight changes in prediabetic patients. evaluate biomarker-based nutrition plans for weight
Environ Med 24: 575--580, 2017. doi:10.5604/ Mol Biol Rep 49: 4217--4224, 2022. doi:10.1007/ loss: the PREVENTOMICS study. Clin Nutr 41: 1834--
12321966.1233555. s11033-022-07254-y. 1844, 2022. doi:10.1016/j.clnu.2022.06.032.

PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org 49

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.


REVIEW
62. Yam P, VerHague M, Albright J, Gertz E, de 77. Bordoni L, Petracci I, Mlodzik-Czyzewska M, 91. Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa
Villena FP, Bennett BJ. Altered macronutrient Malinowska AM, Szwengiel A, Sadowski M, S, Navarrete P, Balamurugan R. The Firmicutes/
composition and genetics influence the complex Gabbianelli R, Chmurzynska A. Mitochondrial DNA Bacteriodes ratio: a relevant marker of gut dysbiosis
transcriptional network associated with adiposity and Epigenetics: Investigating interactions with the in obese patients? Nutrient 12: 1474, 2020.
in the Collaborative Cross. Genes Nutr 17: 13, one-carbon metabolism in obesity. Oxid Med Cell doi:10.3390/nu12051474.
2022. doi:10.1186/s12263-022-00714-x. Longev 2022: 9171684, 2022. doi:10.1155/2022/
9171684. 92. Depommier C, Van Hul M, Everard A, Delzenne NM,
63. Madkour MI, Malhab LJ, Abdel-Rahman WM, de Vos WM, Cani PD. Pasteurized Akkermansia
Abdelrahim DN, Saber-Ayad M, Faris ME. Ramadan 78. Izquierdo AG, Carreira MC, Amil M, Mosteiro CS, muciniphila increases whole-body energy expendi-
diurnal intermittent fasting is associated with atte- Garcia-Caballero T, Fernandez-Quintela A, Portillo ture and fecal energy excretion in diet-induced
nuated FTO gene expression in subjects with MP, Casanueva FF, Crujeiras AB. An energy restric- obese mice. Gut Microbes 11: 1231--1245, 2020.
overweight and obesity: a prospective cohort tion-based weight loss intervention is able to doi:10.1080/19490976.2020.1737307.
study. Front Nutr 8: 741811, 2021. doi:10.3389/ reverse the effects of obesity on the expression of
fnut.2021.741811. liver tumor-promoting genes. FASEB J 34: 2312-- 93. Depommier C, Everard A, Druart C, Plovier H,
2325, 2020. doi:10.1096/fj.201901147RR. Van Hul M, Vieira-Silva S, Falony G, Raes J,
64. Aristizabal MJ, Anreiter I, Halldorsdottir T, Odgers Maiter D, Delzenne NM, de Barsy M, Loumaye A,
CL, McDade TW, Goldenberg A, Mostafavi S, Kobor 79. O’Brien J, Hayder H, Zayed Y, Peng C. Overview of Hermans MP, Thissen JP, de Vos WM, Cani PD.
MS, Binder EB, Sokolowski MB, O’Donnell KJ. microRNA biogenesis, mechanisms of actiones, Supplementation with Akkermansia muciniphila
Biological embedding of experience: a primer on and circulation. Front Endocrinol 9: 402, 2018. in overweight and obese humans volunteers: a
epigenetics. Proc Natl Acad Sci U S A 117: 23261-- doi:10. proof-of-concept exploratory study. Nat Med
23269, 2020. doi:10.1073/pnas.1820838116. 3389/fendo.2018.00402. 25: 1096--1103, 2019. doi:10.1038/s41591-019-
0495-2.
65. Gallardo-Escribano C, Buonaiuto V, Ruiz-Moreno 80. Landrier JF, Derghal A, Mounien L. MicroRNAs in
MI, Vargas-Candela A, Vilches-Perez A, Benitez- obesity and related metabolic disorders. Cells 8: 94. Palmas V, Pisanu S, Madau V, Casula E, Deledda
Porres J, Romance-Garcia AR, Ruiz-Moreno A, 859, 2019. doi:10.3390/cells8080859. A, Cusano R, Uva P, Vascellari S, Loviselli A,
Gomez-Huelgas R, Bernal-Lopez MR. Epigenetic Manzin A, Velluzzi F. Gut microbiota markers
approach in obesity: DNA methylation in a prepu- 81. Formichi C, Nigi L, Grieco GE, Maccora C, Fignani D, associated with obesity and overweight in Italian
bertal population which underwent a lifestyle modi- Brusco N, Licata G, Sebastiani G, Dotta F. Non-cod- adults. Sci Rep 11: 5532, 2021. doi:10.1038/
fication. Clin Epigenetics 12: 144, 2020. doi:10.1186/ ing RNAs: novel players in insulin resistance and s41598-021-84928-w.
s13148-020-00935-0. related diseases. Int J Mol Sci 22: 7716, 2021. 95. Peters BA, Shapiro JA, Church TR, Miller G, Trinh-
66. Kaspar D, Hastreiter S, Irmler M, Hrabe D, Angelis doi:10.3390/ijms22147716. Shevrin C, Yuen E, Friedlander C, Hayes RB, Ahn
M, Beckers J. Nutrition and its role in epigenetic 82. Lauria F, Iacomino G, Russo P, Venezia A, Marena JA. Taxonomic signature of obesity in a large study
inheritance of obesity and diabetes across gener- P, Ahrens W, De Henauw S, Eiben G, Foraita R, of American adults. Sci Rep 8: 9749, 2018.
ations. Mamm Genome 31: 119--133, 2020. doi:10. Hebestreit A, Kourides Y, Molnar D, Moreno LA, doi:10.1038/s41598-018-28126-1.
1007/s00335-020-09839-z. Veidebaum T, Siani A, I.Family Consortium. 96. Koh A, Backhed F. From association to causality:
67. Landecker H. Food as exposure: Nutritional epige- Circulating miRNAs are associated with inflamma- the role of the gut microbiota and its functional
netics and the new metabolism. Biosocieties 6: tion biomarkers in children with overweight and products on host metabolism. Mol Cell 78: 584--
167--194, 2011. doi:10.1057/biosoc.2011.1. obesity: results of the I.Family Study. Genes (Basel) 596, 2020. doi:10.1016/j.molcel.2020.03.005.
13: 632, 2022. doi:10.3390/genes13040632.
68. Li Y. Epigenetic mechanisms link maternal diets and 97. Sharma M, Li Y, Stoll ML, Tollefsbol TO. The epige-
gut microbiome to obesity in the offspring. Front 83. Cantaro G, Filardi T, Sabato C, Vacca A, Migliaccio netic connection between the gut microbiome in
Genet 9: 342, 2018. doi:10.3389/fgene.2018.00342. S, Morano S, Ferretti E. Tissue and circulating obesity and diabetes. Front Genet 10: 1329, 2019.
microRNAs as biomarkers of response to obesity doi:10.3389/fgene.2019.01329.
69. Patti ME. Epigenetic mediators of risk for metabolic treatment strategies. J Endocrinol Invest 44: 1159--
disease across generations. FASEB J 36: 2022. 1174, 2021. doi:10.1007/s40618-020-01453-9. 98. Woo V, Alenghat T. Epigenetic regulation by gut
microbiota. Gut Microbiobes 14: 2022407, 2022.
70. Sasaki A, Murphy KE, Briollais L, McGowan PO, 84. Davis CD. The gut microbiome and its role in obe- doi:10.1080/19490976.2021.2022407.
Matthews SG. DNA methylation profiles in the blood sity. Nutr Today 51: 167--174, 2016. doi:10.1097/
of newborn term infants to mothers with obesity. NT.0000000000000167. 99. Connell J, Ho C, Shen N, Moura P, Cai Y, Tanton D,
PLoS One 17: e0267946, 2022. doi:10.1371/journal. Banavar G, Vuyisich M. Evidence-based precision
pone.0267946. 85. Aoun A, Darwish F, Hamod N. The influence of the nutrition improves clinical outcomes by analyzing
gut microbiome on obesity in adults and the role of human and microbial molecular data with artificial
71. Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang probiotics, prebiotics, and synbiotics for weight intelligence (Preprint). bioRxiv 2021.04.24.441290,
T, Wang X. Maternal high-fat diet induces DNA meth- loss. Prev Nutr Food Sci 25: 113--123, 2020. doi:10. 2021. doi:10.1101/2021.04.24.441290.
ylation changes that contribute to glucose intoler- 3746/pnf.2020.25.2.113.
ance in offspring. Front Endocrinol (Lausanne) 10: 100. Lee YC, Christensen JJ, Parnell LD, Smith CE,
871, 2019. doi:10.3389/fendo.2019.00871. 86. Dibaise JK, Zhang H, Crowell MD, Krajmalnik-Brown Shao J, McKeown NM, Ordovas JM, Lai CQ.
R, Decker A, Rittmann BE. Gut microbiota and its Using machine learning to predict obesity
72. Schulz L. The Dutch Hunger Winter and the devel- possible relationship with obesity. Mayo Clin Proc based on genome-wide and epigenome-wide,
opmental origins of health and disease. Proc Natl 83: 460--469, 2008. doi:10.4065/83.4.460. gene-gene and gene-diet interactions. Front
Acad Sci U S A 107: 16757--16758, 2010. Genet 12: 783845, 2021. doi:10.3389/fgene.
doi:10.1073/pnas.1012911107. 87. Newman TM, Shively CA, Register TC, Appt SE, 2021.783845.
Yadav H, Colwell RR, Fanelli B, Dadlani M, Graubics
73. Bleker LS, de Rooij SR, Painter RC, Ravelli AC, 101. National Institutes of Health. NIH awards $170 million
K, Nguyen UT, Ramamoorthy S, Uberseder B, Clear
Roseboom TJ. Cohort profile: the Dutch famine for precision nutrition study (Online) Washington, DC:
KY, Wilson AS, Reeves KD, Chappell MC, Tooze JA,
birth cohort (DFBC)-a prospective birth cohort study NIH, 2022. https://www.nih.gov/news-events/news-
Cook KL. Diet, obesity, and the gut microbiome as
in the Netherlands. BMJ Open 11: e042078, 2021. releases/nih-awards-170-million-precision-nutrition-
determinants modulating metabolic outcomes in a
doi:10.1136/bmjopen-2020-042078. study#::text=The%20National%20Institutes%20of
non-human primate model. Microbiome 9: 100,
74. Veenendaal MV, Painter RC, de Rooij SR, Bossuyt 2021. doi:10.1186/s40168-021-01069-y. %20Health.
PM, van der Post JA, Gluckman PD, Hanson MA,
88. Nie X, Chen J, Ma X, Ni Y, Shen Y, Yu H, Panagiotou 102. Centers for Disease Control and Prevention. Pre-
Roseboom TJ. Transgenerational effects of prenatal
G, Bao Y. A metagenome-wide association study of valence of obesity and severe obesity among
exposure to the 1944-45 Dutch famine. BJOG 120:
gut microbiome and visceral fat accumulation. adults: United States, 2017--2018 (Online). NCHS
548--553, 2013. doi:10.1111/1471-0528.12136.
Comput Struct Biotechnol J 18: 2596--2609, 2020. Data Brief No. 360, 2020. https://www.cdc.gov/nchs/
75. Meng R, Lv J, Yu C, Bian Z, Yang L, Chen Y, Zhang doi:10.1016/j.csbj.2020.09.026. products/databriefs/db360.htm#::text=Examination%
H, Chen X, Chen J, Chen Z, Chen J, Chen Z, Qi L, lLi 20Survey%20.
L, China Kadoorie Biobank Collaborative Group. 89. Rampelli S, Guenther K, Turroni S, Wolters M,
Prenatal famine exposure, adulthood obesity pat- Veidebaum T, Kourides Y, Molnár D, Lissner L, 103. Choi WJ, Shin D. Interactions between red and
terns and risk of type 2 diabetes. Int J Epidemiol 47: Benitez-Paez A, Sanz Y, Fraterman A, Michels N, processed meat consumption and APOA5 gene
399--408, 2018. doi:10.1093/ije/dyx228. Brigidi P, Candela M, Ahrens W. Pre-obese child- variants associated with the incidence of metabolic
ren’s dysbiotic gut microbiome and unhealthy diets syndrome in Korean adults. Genes Nutr 17: 5, 2022.
76. Crujeiras AB, Izquierdo AG, Primo D, Milagro FI, may predict the development of obesity. Commun doi:10.1186/s12263-022-00707-w.
Sajoux I, Jacome A, Fernandez-Quintela A, Portillo Biol 1: 222, 2018. doi:10.1038/s42003-018-0221-5.
MP, Martinez JA, Martinez-Olmos MA, de Luis D, 104. Rask-Andersen M, Karlsson T, Ek WE, Johansson Å.
Casanueva FF. Epigenetic landscape in blood leu- 90. Yan H, Qin Q, Chen J, Yan S, Li T, Gao X, Yang Y, Li Gene-environment interaction study for BMI reveals
cocytes following ketosis and weight loss induced A, Ding S. Gut microbiome alterations in patients interactions between genetic factors and physical
by a very low calorie ketogenic diet (VLCKD) in with visceral obesity based on quantitative com- activity, alcohol consumption and socioeconomic
patients with obesity. Clin Nutr 40: 3959--3972, puted tomography. Front Cell Infect Microbiol 11: status. PLoS Genet 13: e10069770, 2017. doi:10.
2021. doi:10.1016/j.clnu.2021.05.010. 823262, 2022. doi:10.3389/fcimb.2021.823262. 1371/journal.pgen.1006977.

50 PHYSIOLOGY  Volume 38  January 2023  www.physiologyonline.org

Downloaded from journals.physiology.org/journal/physiologyonline (187.019.178.241) on March 10, 2024.

You might also like