Skip to main content
Advances in Nutrition logoLink to Advances in Nutrition
. 2021 Apr 22;12(5):2023–2034. doi: 10.1093/advances/nmab040

Synergizing Mouse and Human Studies to Understand the Heterogeneity of Obesity

Penny Gordon-Larsen 1,2,, John E French 3,4, Naima Moustaid-Moussa 5, Venkata S Voruganti 6,7, Elizabeth J Mayer-Davis 8, Christopher A Bizon 9, Zhiyong Cheng 10, Delisha A Stewart 11,12, John W Easterbrook 13, Saame Raza Shaikh 14,
PMCID: PMC8483969  PMID: 33885739

Abstract

Obesity is routinely considered as a single disease state, which drives a “one-size-fits-all” approach to treatment. We recently convened the first annual University of North Carolina Interdisciplinary Nutrition Sciences Symposium to discuss the heterogeneity of obesity and the need for translational science to advance understanding of this heterogeneity. The symposium aimed to advance scientific rigor in translational studies from animal to human models with the goal of identifying underlying mechanisms and treatments. In this review, we discuss fundamental gaps in knowledge of the heterogeneity of obesity ranging from cellular to population perspectives. We also advocate approaches to overcoming limitations in the field. Examples include the use of contemporary mouse genetic reference population models such as the Collaborative Cross and Diversity Outbred mice that effectively model human genetic diversity and the use of translational models that integrate -omics and computational approaches from pre-clinical to clinical models of obesity. Finally, we suggest best scientific practices to ensure strong rigor that will allow investigators to delineate the sources of heterogeneity in the population with obesity. Collectively, we propose that it is critical to think of obesity as a heterogeneous disease with complex mechanisms and etiologies, requiring unique prevention and treatment strategies tailored to the individual.

Keywords: heterogeneity, obesities, mouse, human, pre-clinical, clinical, prevention, treatment, nutrition, symposium

Introduction

Like cancer decades ago, obesity is considered by many as a single disease state with universal treatment, rather than as a fundamentally heterogeneous process varying in mechanisms and etiologies, each requiring unique prevention and treatment strategies. This “one-size-fits-all” approach has not served patients or communities well; obesity and its complications continue to rise with significant health-care burden. This article, the result of the first annual University of North Carolina Interdisciplinary Nutrition Sciences Symposium, focuses on a set of factors important to translational science in studies aiming to synergize human and animal models of obesity heterogeneity. In addition, we provide suggestions for best practices to delineate the sources of heterogeneity that will ultimately lead to a better understanding of underlying mechanisms and precision medicine/precision nutrition treatment approaches.

Obesity is Highly Heterogeneous with Many Underlying Sources

Obesity is defined as excess body fat. The most common clinical practice for adults is to use body mass index [BMI; wt (kg)/ht (m)2] to screen and diagnose overweight/obesity to identify cardiometabolic risks (1). However, it is important to recognize that the BMI does not distinguish lean and fat body compartments, as such BMI is problematic on its own for clinical use at the individual level for estimating body fat and lean mass (2). In addition, at any given BMI, there is heterogeneity in body fat distribution as well as differential association with cardiometabolic disease risk (1). For this reason, recent clinical guidance suggests including measures of waist circumference at given BMI values to improve risk stratification across age, sex, and ethnicity (3). Indeed, The International Diabetes Federation recommends sex- and ethnic-specific waist circumference cut points (e.g., for Asian populations) to allow for the differential risk across populations (4).

The complex disease of obesity results from a range of individual predisposition factors (including genetic, epigenetic, biologic, hormonal, microbial, early life events) as well as a range of environment (geography, nutritional status, contaminant exposures) and lifestyle factors (including built/physical environment, cues/social habits, food cost/availability, taste/smell/palatability) (5, 6). Individual predispositions shape responses to environment and lifestyle factors; derangements in this system lead to obesity. This complex multifactorial etiology makes it challenging to identify the mechanisms and causes of obesity heterogeneity. In addition, there is a paucity of well-characterized, population representative datasets that have information on individual predisposition factors as well as the range of obesity-relevant environment and lifestyle factors. Furthermore, the computational and methodological complexity of analyzing multi-omic and multilevel data in large population datasets cannot be understated.

It is beyond the scope of this review to discuss the intricate details of each source of heterogeneity in the population with obesity (7–9). Herein, we focus on key factors that are critical in synergizing human and animal models of obesity heterogeneity, which are summarized in Table 1. One notable factor in the heterogeneity of obesity is sex. At the same BMI, females tend to have more body fat than men. Body composition tends to be sexually dimorphic with central adiposity a strong indicator of cardiometabolic risk. People with a pear shape tend to carry weight in the hip area, while people with an apple shape tend to have excess fat in the abdominal area, a more cardiometabolically adverse patterning due to metabolically active adipose as energy metabolism and endocrine functions vary with locations of fat deposition (10). There is suggestion that central adiposity may confer higher cardiovascular risk among women than men (11). Understanding such sex differences may shed light on the pathophysiology of adiposity and offer insight for potential interventions aimed at women versus men.

TABLE 1.

Potential sources of heterogeneity in obesity that should be further integrated between rodent and human studies

Sources of heterogeneity Description
Sex • Sex differences are well established in human and rodent metabolism in the context of obesity. However, more work is needed in integrating sex-specific studies, particularly at the rodent level, with human studies.
Race/ethnic background • It is critical to incorporate race/ethnic differences in human studies as they give rise to heterogeneity in the human population. These results can then guide mechanistic experiments at the rodent level.
Age at onset of obesity • Rodent studies that compare outcomes with humans need to account for differences in the age of onset of obesity.• Furthermore, comparing data within the human population also needs to account for the age of onset and duration of obesity.
Genetic background • Host genetics are a well-established source of heterogeneity in the human population. Pre-clinical studies could increasingly incorporate the use of CC and DO mouse models to better understand the role of host genetics in obesity heterogeneity. The next steps will be to integrate these data with human studies.
Tissue/cellular heterogeneity • As an example, adipose tissue depots are not uniform within a human or a mouse. Thus, dissecting the role of specific adipose tissue depots on metabolic outcomes is of significance.• The abundance and function of differing cell types (e.g., adipocytes, immune cells) within a given tissue can vary considerably. Thus, there is a need to understand how heterogeneity at the tissue and cellular levels gives rise to variation in humans with obesity and rodent models.

CC, collaborative cross; DO, diversity outbred.

Even within a given sex, there are additional factors that must be considered such as race. For example, gay, lesbian, or gender queer adults may have differential patterns of cardiovascular risk with obesity (12). Further, there are established differences in BMI cut points for Asians given higher cardiovascular risk at lower BMI (4). Similar differences are likely for other race/ethnic groups (13–15) and may result from a combination of biological and structural societal factors such as racism (16). As one example, gluconeogenesis in premenopausal black women is lower than in white women, which has strong implications for diagnosis of pre-diabetes (17). This research gap is increasingly being addressed with a recent requirement, when possible, by the NIH for inclusion of women and populations underrepresented in research involving human subjects and the requirement for addressing sex as a biological variable in research using rodent models.

Besides sex, race, and ethnicity, age at onset of obesity may give rise to obesity heterogeneity. Childhood obesity is associated with increased risk of numerous complications, including but not limited to type 2 diabetes, malignancies, autoimmunity, psychiatric problems, reproductive complications, etc. (18, 19). Of course, more work is needed in this area of research as there is also some discrepancy in the field as many have failed to disentangle duration of obesity with age at onset or have not adequately addressed the complex multifactorial nature of obesity. For instance, one study that focused on adult candidates for bariatric surgery showed that lower age at onset of obesity predicted higher BMIs; however, these same individuals were less prone to hypertension and type 2 diabetes compared with those with adult-onset obesity (18). Nevertheless, the age at onset, duration of obesity, and other complex factors are critical research gaps to consider in studies of obesity and its complications from the pre-clinical to clinical level. Furthermore, when comparing mouse and human data, the age at onset of obesity may be a factor that is routinely ignored and could be a factor that can improve synergizing mouse and human studies of obesity (20).

Genetics is a major source of heterogeneity in the human population. Most genetic studies of obesity susceptibility have failed to utilize quantitative diet data to interrogate gene variations in obesity that may only be revealed when considering diet. For example, a gene polymorphism that decreases thermogenesis (i.e., rate at which calories are burned) may be of no consequence in people with a low-calorie diet but will influence weight in high calorie consumers. In studies that ignore differential exposures (e.g., pooling responders and non-responders), gene susceptibility is missed. Similarly, few obesity interventions are tailored to individual susceptibility, even to well-established susceptibility factors such as glycemic status. Furthermore, few collect genetic, metabolomic, or microbial data, prohibiting investigation of differential treatment effects and identification and characterization of underlying biologic pathways. We discuss these issues in greater detail below on how to potentially bridge this gap with the integration of newer mouse models for obesity and systems approaches for human research.

While obesity is typically associated with metabolic abnormalities and cardiometabolic diseases, there is individual variation in this risk with differences in patterning of disease risk across obesity, including some individuals with obesity with few cardiometabolic complications. It is well known that obesity perturbs metabolic pathways (21), thereby affecting cardiovascular disease (CVD) risk factors (e.g., cholesterol, blood pressure, and glycemic phenotypes) and their sequelae (22–28) as well as heterogeneity in association with a range of other diseases, from cancers (29) to infections (30). In fact there are many papers classifying people with metabolically healthy obesity, albeit with a large range in definitions and classifications (31). Yet it is important to note that a range of modifiable lifestyle factors, adipose tissue biology, or differential mechanisms (in addition to methodological differences in classification and temporal effects) may underlie differences in metabolic health within the population with obesity (32–35). These differences can point to subtypes of obesity and shed light on mechanisms underlying the heterogeneity of obesity.

We know little about exactly how obesity stresses metabolic pathways during younger adulthood when CVD risk accelerates; the specific biologic mechanisms remain poorly understood (36, 37). Such research gaps reflect several challenges, including a preponderance of studies evaluating lifecycle period after CVD is established (38–42). There is a clear need to better understand the evolution of CVD in the context of unremitting metabolic stress induced by obesity (43, 44) and CVD risk factors (45–48). The “expressed genome”—factors beyond DNA such as epigenomics and metabolomics—offers innovative opportunities to fill this major research gap (49–52).

Tissue and Cellular Heterogeneity May Drive Heterogeneity in the Population with Obesity

We have addressed several issues such as sex, race, ethnicity, age at onset of obesity, and genetics as they relate to heterogeneity at the population level. However, heterogeneity of obesity is often ignored at the tissue and cellular level in pre-clinical- and population-level studies. Here we use adipose tissue as a case in point (53, 54). Adipose tissue presents several levels of heterogeneity with distinct properties and functions, especially in white adipose tissue (WAT) (53), as illustrated by 1) different types of adipose tissues known as brown, white, and beige adipose tissues, with different locations within the body, and 2) cellular heterogeneity in cell types and cell size, as adipose tissue is composed of several cell types, including pre-adipocytes, stem cells, immune cells, and adipocytes among other cells.

WAT is a primary storage organ of triglycerides during energy excess. WAT influences systemic metabolism not only through availability of these stores that can be released as fatty acids when needed, but also through secretion of numerous hormones and adipokines secreted by adipose tissue (55). In contrast, brown adipose tissue (BAT) is a major driver of thermogenesis and energy expenditure through a specialized mitochondrial protein—uncoupling protein 1 (UCP-1)—and heat generation in both humans and rodents (56, 57). Over the past decade, beige adipose tissue emerged as a third type of adipose tissue. Beige adipocytes are brown like adipocytes and positive for UCP-1 and arise within white fat (also named brite for “brown in white”). Published research shows that these tissues not only differ in metabolic functions, but they also exhibit distinct molecular differences (58).

Adipose tissue expands through hyperplasia (proliferation then differentiation of adipose stem cells or pre-adipocytes) and/or through hypertrophy (increased mature adipocyte cell size). The latter, especially for WAT, has significant implications on obesity-related diseases. Indeed, the inability of adipocytes to expand and continue storing energy leads in part to “spillover” of lipids into non-adipose tissues such as liver, muscle, and pancreas, causing lipotoxicity and associated metabolic dysfunctions, including insulin resistance (59, 60). Moreover, adipocytes come in different sizes (61), and it is generally recognized that smaller adipocytes are associated with insulin-sensitive phenotypes while large adipocytes are associated with insulin resistance. This has been well illustrated in several animal models—specifically, insulin receptor knockout models as well as angiotensinogen and angiotensin II receptor transgenic/knockout models (62, 63).

It is important to recognize that differing cell types, notably adipocytes, found in various adipose tissue depots are developmentally distinct. Thus, differences in fat distribution across depots are driven, in part, by the ability of each unique progenitor cell to grow and differentiate. This developmental programming is under the control of a unique transcriptome, which is likely further regulated by sex hormones (58). Furthermore, there is a strong appreciation for heterogeneity in numerous immune cell populations, each with their own specialized metabolic profile, within differing WAT depots that control the inflammatory tone (64).

Understanding the aforementioned heterogeneity of adipose tissues and adipose tissue distribution, in addition to the heterogeneity of adipocytes and immune cells, using mouse models and well-defined human fat samples may help stratify the different types of “obesities,” which will ultimately result in designing better targeted interventions for metabolic diseases. Specifically, given the limited amount of BAT in humans, a promising target is WAT, including understanding mechanisms and interventions that can reduce it and/or increase potential conversion of white into brite adipocytes. Moreover, development of animal models that better mirror heterogeneity in metabolism and distribution of adipose tissues may shed light on how adipose tissues impact whole body metabolism and lead to different metabolic outcomes of obesity.

Collaborative Cross and Diversity Outbred Mouse Populations Are Powerful for Modeling Complex Diseases and May Increase Understanding of the Heterogeneity of Obesity

Diverse populations are key to understanding heterogeneity in obesity. Our major breakthrough in understanding the genetic underpinnings of obesity's complexity have come from large, population-based genetic consortia (65–75). Yet even among these large studies, there is generally a historic gap in studies in multiethnic human populations, with most work in European Americans (76–78). This is a problem because of poor transferability of European-American trait-associated variants to multiethnic populations (79, 80). Of course, new data are starting to emerge from multiethnic populations, which are critical for investigating the heterogeneity of obesity. As one example, a genome-wide association study (GWAS) from 100,418 adults from the multiethnic Genetic Epidemiology Research on Adult Health and Aging (GERA) cohort identified 30 novel BMI autosomal loci (81). For instance, KDM4C was identified, which is a transcription factor involved in regulating adipogenesis (81). Some of these data are publicly available and are a rich resource for further investigation (82).

Most animal studies have capitalized upon standardized genomic backgrounds that are homogeneous (homozygous inbred). Further, animal studies have typically only interrogated variation at a single locus. Thus, these types of studies provide a poor model for human variation relative to complex multigenic diseases, like obesity. In contrast, novel heterogeneous mouse populations, like the collaborative cross (CC) and diversity outbred (DO), better approximate human populations in terms of genetic diversity and thus are excellent for investigating complex disease biology (83).

Homozygous inbred lines of mice have been a useful pre-clinical experimental research model with success in basic and biomedical research with qualifications (84). To aid quantitative research similar to human GWAS, new mouse models and approaches have been created and used for genetic analysis (85–90). Hybrid mouse diversity panels, the collaborative cross recombinant inbred lines (CCRIL) and DO mice, are representative of haplotype association mapping and linkage analysis approaches in pre-clinical experimental population-based mouse models. Hybrid mouse diversity panels are a powerful tool with more than 8 million single nucleotide polymorphisms (SNPs) but are limited by significant intervals that are identical by descent (IBD) in some or all laboratory-derived homozygous inbred lines (91, 92). This approach may be limited for robust quantitative trait loci (QTL) analysis for some phenotypes.

The CCRIL and the DO mice have different strengths and potential limitations. The CCRIL mice were created through a multiple advanced generation intercross using 8 homozygous inbred lines of mice selected based on particular attributes (93). The DO mice were created from early progenitors of the established CCRIL in 2 steps starting from a founding population of 150 sister–brother pairs of partially inbred CC lines sampled at the sixth filial generation from the second generation (G2F6) (94). Simulated rounds of mating in which females and males were paired at random with the constraint that sib-mating was prohibited were used (94). The final inbred lines of the CCRIL and the randomized DO mating project contain approximately 45 million single nucleotide polymorphic and structural variants and a 12% minor allele frequency (MAF). Together, they represent powerful tools for quantitative genetics and identifying QTL based on the variance contributed by one or more founder haplotype of origin (95, 96). The 8 founder lines contributed significant genetic diversity and a high MAF for genetics to aid quantitative trait analysis to identify genes, and bioinformatic analysis to explain significant variance associated with phenotype. These discovery tools further enable reverse genetics studies to demonstrate causal relationships using the founder lines or the CCRIL. The genetic sequence and identification of phenotype-associated QTL identified candidate gene SNP or structural variants and tools to aid mouse to human translation are available on the Mouse Genome Informatics database at The Jackson Laboratories (97, 98). For further information, please refer to The Jackson Laboratory Mouse Genome Informatics (MGI; http://www.informatics.jax.org) and Mouse Phenome Database (MPD; https://phenome.jax.org/).

It is important to note that the creation of loss and/or gain of function mutants based on “single locus” of origin (homozygous inbred strain) used in genetically altered mouse models for pre-clinical research does not apply to the CCRIL and DO mice models. Each CCRIL or DO mouse has 8 different alleles (haplotype) at each genetic locus. Each CCRIL is isogenic (>95% homozygosity at each locus) allowing for co-isogenic controls in experimental balanced block design.

Many homozygous inbred strains derived in the laboratory, principally from the Mus musculus domesticus subspecies, have significant IBD. IBD limits genetic diversity at specific loci and significantly decreases the statistical ability to identify phenotype-specific protocol-driven QTLs based on non-synonymous SNPs and/or copy number variations. Thus, our understanding and accounting for the multigenic basis of the phenotypic trait to explain phenotype variance can be decreased. Depending upon study design, the randomly bred genetically diverse DO mice and the isogenic inbred CCRILs capture a similar magnitude of genetic diversity. Together, they provide increased opportunities to develop methods necessary to extrapolate between these genetically diverse mouse models and humans based on similar phenotype and genotype.

Weight gain and loss risk variants identified in DO mice studies can be tested in selected CCRIL lines mice with CRISPR/Cas9 modifications to investigate specific allelic variants and their mechanisms to demonstrate proof of causality. For example, population-based genetically diverse mouse models can be used to dissect complex traits related to nutrient overload (83, 99). Several studies in CCRIL and DO mice have demonstrated the use of these population-based models to identify QTLs for explaining human phenotypic variation (100–103).

Taken together, we consider the randomized DO mice as forward genetics or discovery models and the isogenic CCRILs for corroborating phenotypes and QTLs leading to hypothesis-based research with isogenic controls as well as traditional comparisons of phenotypic responder and non-responder F1 and F2 outcrosses. For instance, DO mice are being used as a discovery model to determine why some obese mice have improved hyperinsulinemia and hyperglycemia with select dietary interventions whereas other mice have impaired hyperinsulinemia and hyperglycemia (99). To further exemplify, in our DO mice studies focused on the heterogeneity of obesity based on operational paradigms, we are using series of DO mice cohorts (50 to 100 of each sex) over time to gain power for linkage analysis and discovery of candidates that explain the haplotype(s) of origin and the majority of the operationally defined phenotypic variance (research paradigm dependent). Once the haplotype is identified that explains the phenotypic variance, we can use the CCRIL with and without the genetic locus specific haplotype and retesting, and compare and contrast outcomes (JE French et al., manuscript in preparation). This also allows for use of the appropriate CCRIL for creating haplotype-specific controls and experimental groups to test gene × diet interactions. We are working toward using experimental clinical designs and phenotypic/genetic analysis outcomes and incorporating the elements of those designs based on the features of the CCRIL and DO mouse models to test complementarity between SNP-based GWAS or family linkage analysis approaches.

Integration of -Omics and Computational Approaches to Tackle the Heterogeneity of Obesity

For many decades, obesity research has addressed mechanisms involving 1 single factor (e.g., using a single type of data such as either genetics or metabolomics rather than both genetics and metabolomics) in a single model (e.g., human or single inbred mouse strain) at a single point in time. Yet current advances in computational efficiency, data science, and measurement technologies provide outstanding opportunities for investigating the heterogeneity of obesity. For example, we can now start to integrate a wide range of -omics (i.e., metabolomics, lipidomics, proteomics, genomics, microbiome) into pre-clinical and clinical studies to ultimately establish the underlying mechanisms and potential treatment approaches across different subtypes of obesity. Although this approach has been rarely applied to obesity heterogeneity, there are emerging studies that integrate various -omics analyses in the human population. To exemplify, a recent epidemiological study examined associations between the gut microbiota and the plasma metabolome with blood pressure in a Chinese cohort. The data revealed unique microbiota and metabolite signatures (notably of acyl-carnitines and differing lipids) that were associated with systolic and diastolic blood pressure (104). Additionally, using complex multi-omics data in combination with integrative analysis and systems biology along with expertise in obesity and metabolism has the potential to transform current understanding of the heterogeneity of obesity.

The rise of publicly available structured biomedical knowledge has allowed the creation of large-scale knowledge integration projects such as the NCATS Biomedical Data Translator (https://doi.org/10.1111/cts.12591), including the creation of the ROBOKOP Knowledge Graph (KG) (https://doi.org/10.1021/acs.jcim.9b00683). As an example, the ROBOKOP KG compiles information from a dozen public sources and contains information on over 4 million biomedical entities including genes, diseases, phenotypes, chemicals, anatomical features, and sequence variants as well as over 12 million relationships between these entities. The KG, which is available for browsing and download at http://robokopkg.renci.org, serves as an integration point for observed obesity associations. These associations are loaded into the graph as new relationships, such that subsequent database queries return association data combined with background information, providing the framework for a mechanistic interpretation of new associations focused on the heterogeneity of obesity.

The flexibility of knowledge graphs to integrate heterogenous data also allows a systematic representation of information across species. For instance, the Monarch Initiative Knowledge Graph relates phenotypes, anatomical features, and genes across model organisms so that genotype/phenotype relations observed in, for instance, mice, can be used to suggest or support orthologous relations in humans (105). The structure of the KG, therefore, allows for parallel mechanisms to be explored across species without losing context of the particular organism.

Risk Factor and Genetic Clustering May Reveal Subtypes of Obesity

An additional approach for tackling the identification of differing types of “obesities” will be to cluster differing biomarkers of disease, risk factors, or even genetic pathways. These biomarkers could be well established clinical measures or could even be the identification of new and validated parameters from -omic approaches described earlier. Here we describe a couple of examples from studies focused on the heterogeneity of diabetes.

As an example, a study used k-mean and hierarchical clustering analysis of nearly 9000 newly diagnosed participants with diabetes. The clusters were defined by BMI, HbA1c, age at diagnosis of diabetes, circulating concentration of glutamate decarboxylase antibodies, and homoeostatic model assessment of pancreatic β-cell function and insulin resistance. These parameters were then associated with patient record data, including complications of diabetes and use of prescription drugs. The analyses revealed 5 groups of patients with diabetes, with each cluster having unique characteristics and risks. For instance, one notable finding was that those that were the most insulin resistant had the highest risk for fatty liver and kidney disease (106, 107).

To further exemplify, in another study, GWAS results were clustered using Bayesian non-negative matrix factorization for 94 type 2 diabetic genetic variants and 47 type 2 diabetes traits (108). Analyses from this study revealed 5 genetic clusters with distinct traits that appeared to represent unique mechanistic pathways that drive the onset and/or progression of type 2 diabetes. This approach highlighted the possibility of stratifying individuals based on distinct genetic pathways that could predict physiological outcomes. Taken together, these results underscore the potential utility of these approaches that can be applied to the field of obesity to drive future precision medicine and precision nutrition studies. In addition, establishing underlying cellular and molecular mechanisms with rodent models will aid in our understanding of why some pathways favor specific physiological outcomes.

Best Practices for Animal and Human Research on the Heterogeneity of Obesity

There is a strong need for establishing uniform practices in the study of obesity heterogeneity using pre-clinical and clinical models to ensure rigor and reproducibility. There are several variables that we discuss below that are often ignored in the field. One major variable is the lack of consideration for gene × diet interactions. Obesity and its complications such as type 2 diabetes and CVDs are influenced by multiple genetic and environmental factors, including diet behaviors. Genome-wide association studies are the best means of confirming known and discovering novel variants associated with obesity. Some studies have been successful in identifying genetic variants and their interactions with environmental factors in influencing the disease process and/or risk factors (109–111). However, most of these studies are lagging behind with respect to lifestyle interactions (e.g., sleep, physical activity, stress, smoking, and alcohol consumption), despite known recognition that inclusion of lifestyle intake data reduces the noise in genetic signals (112–114). Studies that have reported and replicated gene–nutrient interactions affecting obesity have mostly focused on macronutrients, mainly fat and carbohydrate intake (115–117), with few focusing on meal patterns. Key genes whose interactions with nutrients/meal patterns have been reported and replicated across studies include apolipoprotein A2 (APOA2), fat mass and obesity associated (FTO), melanocortin 4 receptor (MC4R), lipoprotein lipase (LPL), and peroxisome proliferator activated receptor gamma (PPARγ) (118–122).

Diet can regulate gene expression by affecting transcription (RNA processing and stability), RNA translation, and proteins and metabolite processing. In turn, metabolism of nutrients is affected by the genetic sequence and architecture of the individual (123, 124). Evolutionarily, nutritional environments seem to be the major determinants of human variation, given that populations vary in requirement for foods and response to diet (124). Thus, the fields of nutrition and genetics are intertwined; studies of human or animal genetics are not complete without taking into consideration nutritional variability in the population with obesity. However, diet × nutrient interactions account for a small portion of the variation in obesity. Another component of lifestyle that has evoked interest is the role of gene by physical activity interactions in obesity. Genes such as angiotensin-converting enzyme (ACE), angiotensinogen (AGT), alpha actinin 3 (ACTN3), and FTO have been consistently shown to interact with physical activity and to be associated with adiposity/obesity (125–128). Other studies have reported interactions of genetic variants with other factors such as sleep (129, 130), stress (131), smoking (132, 133), alcohol intake (134), and even socioeconomic status (135–137).

It is important to note that gene by lifestyle interactions in obesity have been mainly examined using genome-wide and candidate gene association studies. Several models have been proposed to estimate and analyze interaction effects with obesity, where most are regression based. Some of the issues that need to be taken into consideration while examining gene by lifestyle interaction effect on obesity include selection of genetic models (additive vs. dominant vs. recessive), interaction models (additive or multiplicative terms), and type of confounders and time of exposure (113, 138).

Another limitation is the differences in the instruments used to measure dietary intake. Dietary intake assessment is usually conducted using food records, 24-h recalls, and FFQs. The ability of food records and 24-h recalls to capture usual intake depends on the number of days assessed. Similarly, the accuracy of FFQs, designed to capture long-term intake patterns, is also affected by the number of days assessed along with the number and relevance of foods included, in addition to the recall bias. All these factors need to be considered and standardized across studies to ensure rigor and reproducibility. One approach that can be integrated into studies is the use of mobile technology and wearable-based detection approaches, which of course have their own challenges (139–141).

Another constraint in advancing the science of obesity heterogeneity is the lack of uniformity in diet composition to achieve an obese phenotype. For example, the fat source for high-fat diets varies routinely in pre-clinical experiments. A range of fat sources are implemented in the experimental diets, including coconut oil, palm oil, lard, milkfat, and mixed oils (142). Similarly, a control diet for high-fat feeding studies is often a purified mouse diet or at times, erroneously, a standard mouse chow (142). However, the ingredients found in a standard mouse chow are highly variable, containing many additional substances such as pesticides, heavy metals, and phytoestrogens (143). It is no wonder that reproducibility of effects or lack of translation from animal to human models is often problematic. Greater attention must be paid to diet in design and interpretation of studies, particularly relevant to consistency in controls across laboratories, to make rigorous conclusions about any given macro- or micronutrient. Like the use of the ARRIVE (Animal Research: Reporting of In Vivo Experiments) checklist and other guidelines for mouse studies (144, 145), we are in need of strong nutrition guidelines for obesity research.

The case in point about control diets also applies to human intervention studies. For instance, what are appropriate controls for human dietary supplement studies? If we stick with studies of dietary fat as a variable, then what are appropriate placebo controls?

Again, a limiting factor is the lack of collection of high-quality diet data in population, cohort, and clinical studies. The few studies that incorporate diet measures rely upon a wide range of methods used to capture dietary intake, which makes cross-study harmonization very difficult. Given the role of diet/nutrients in obesity, there is a need to integrate high-quality nutritional assessment tools and nutritional biomarkers in population, cohort, and clinical studies, and to use appropriate experimental models to attain proper translation to human conditions.

Understanding obesity as a heterogeneous disease with complex mechanisms and etiologies can take us further to unique prevention and treatment strategies tailored to the individual. In this paper, we have provided some fundamental gaps in knowledge of the heterogeneity of obesity ranging from the cellular level (e.g., heterogeneity of adipocytes and immune cells across fat depots) to population perspectives (age at onset of obesity, sex, host genetics). It was not our intent to address the full scope of individual predisposition factors and the full range of environment and lifestyle factors. We specifically focus on a set of factors important to translational science in synthesizing human and animal models of obesity heterogeneity. As such, we are not including a wide range of other factors that are important but may relate only to human or only to mouse studies. Taken together, we propose the following measures to improve studies of obesity heterogeneity and to enhance translation from mouse to human:

  1. Build translational teams to address heterogeneity of obesity. Integrating expertise across basic, computational, clinical, and population for true translational science and integrative analyses is critical to making headway in understanding the heterogeneity of obesity. Application of -omic (i.e., metabolomics, lipidomics, proteomics, genomics, microbiome) and computational approaches will also be key.

  2. Analyses of validated SNPs that could account for differences in nutrient metabolism. This will take us one step closer to addressing the neglect of gene × diet interactions. Furthermore, increased utilization of population-based models such as DO and CC mice as models for complex diseases, will provide key information about novel SNPs in the heterogenous population with obesity.

  3. Experimental diets for rodent and human studies should undergo rigorous quality control analyses by investigators prior to and during the course of a study and the results should be reported.

  4. The bulk of current studies that rely on 45% and 60% kcal from fat diets do not model human intake of carbohydrates and fats. Investigators at the pre-clinical level should start the use of newly emerging obesogenic diets that model human macronutrient intake (146).

  5. Account for sex differences in human and mouse studies. While there is increasing appreciation for sex differences, this area of research needs to be further expanded as sex differences will contribute toward differences in various measured outcomes. Notably, rodent studies are still heavily focused on male mice although there is a greater appreciation for conducting experiments with females based on a recent push from the NIH. These studies will need to address sex differences from the population level to differences in underlying mechanisms of action.

  6. Account for the age at onset and duration of obesity. This is particularly relevant when comparing results between mice and humans.

  7. Incorporate race/ethnic diversity in human studies as an important source of heterogeneity and a marker for structural factors that contribute to disease.

Conclusion

Collectively, there is a critical need to understand the underlying sources of heterogeneity in the population with obesity. Translational studies spanning mouse and human populations offer one such direction of research. Sources of heterogeneity range from sex/race difference, age at onset of obesity, differing genetic backgrounds, diversity in the diet, variations across individuals in their -omic profiles (microbiome, metabolome, lipidome, genome, proteome), and differences among individuals in their underlying cellular profiles within key tissue depots such as brown and white adipose tissue. Addressing the underlying causes of heterogeneity in obesity and developing precision medicine and precision nutrition treatment approaches will rely on large-scale integration of the next generation of population-based mouse models with human clinical studies. Finally, there is a need to develop better practices that will allow for strong rigor and reproducibility in the study of obesity heterogeneity.

ACKNOWLEDGEMENTS

The authors acknowledge the keynote speakers at the first annual Interdisciplinary Nutrition Sciences Symposium held at the University of North Carolina at Chapel Hill on 24–25 July, 2019: J Carl Barrett, Gary Churchill, Lee M Kaplan, Francine Kaufman, Fernando Pardo-Manuel de Villena, and Steven R Smith. The authors' responsibilities were as follows—All authors contributed toward the writing of the article; PG-L and SRS also proofread and edited the article; PG-L and SRS assume responsibility for the entire review; and all authors: have read and approved the final manuscript.

Notes

The authors acknowledge funding support from the following sources: R13DK122823 and R01HL143885 (PG-L), UNC-CH OVCR Creativity Hubs Pilot Award (PG-L, JEF, CAB), R15AT008879, USDA NIFA 2018-67012-27977, and 19AIREA34450279 (NM-M), R35CA197627 and R21OH011562 (JEF), P30DK056350 (EJM-D, JWE, SRS), OT2TR002514 (CAB), American Heart Association 18TPA34230082 (ZC), R35CA197627 (DAS), R01AT008375 and R03AI59308 (SRS).

Author disclosures: PG-L, JEF, NM-M, VSV, EJM-D, CAB, ZC, DAS, and JWE, no conflicts of interest. SRS is currently supported by Organic Technologies and has previously received support from GSK, Stealth Biotherapeutics, and Methylation Sciences.

Abbreviations used: BAT, brown adipose tissue; CC, collaborative cross; CCRIL, collaborative cross recombinant inbred lines; DO, diversity outbred; GWAS, genome-wide association studies; IBD, identical by descent; MAF, minor allele frequency; KG, ROBOKOP Knowledge Graph; QTL, quantitative trait loci; SNP, single nucleotide polymorphism; UCP-1, uncoupling protein 1; WAT, white adipose tissue.

Contributor Information

Penny Gordon-Larsen, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.

John E French, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA.

Naima Moustaid-Moussa, Obesity Research Institute and Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.

Venkata S Voruganti, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA.

Elizabeth J Mayer-Davis, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.

Christopher A Bizon, Renaissance Computing Institute, University of North Carolina at Chapel Hill, NC, USA.

Zhiyong Cheng, Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA.

Delisha A Stewart, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA.

John W Easterbrook, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.

Saame Raza Shaikh, Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.

References

  • 1.Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA, Hu FB, Hubbard VS, Jakicic JM, Kushner RFet al. . 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014;129(25 Suppl 2):S102–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gonzalez MC, Correia M, Heymsfield SB. A requiem for BMI in the clinical setting. Curr Opin Clin Nutr Metab Care. 2017;20(5):314–21. [DOI] [PubMed] [Google Scholar]
  • 3.Ross R, Neeland IJ, Yamashita S, Shai I, Seidell J, Magni P, Santos RD, Arsenault B, Cuevas A, Hu FBet al. . Waist circumference as a vital sign in clinical practice: a Consensus Statement from the IAS and ICCR Working Group on Visceral Obesity. Nat Rev Endocrinol. 2020;16(3):177–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.WHO Expert Consultation . Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet North Am Ed. 2004;363(9403):157–63. [DOI] [PubMed] [Google Scholar]
  • 5.Gordon-Larsen P, Heymsfield SB. Obesity as a disease, not a behavior. Circulation. 2018;137(15):1543–5. [DOI] [PubMed] [Google Scholar]
  • 6.Lee A, Cardel M, Donahoo WT.. Social and environmental factors influencing obesity. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan Ket al., editors. Endotext. [Internet]. South Dartmouth, MA: MDText.com, Inc; 2000. [Google Scholar]
  • 7.Brandão I, Martins MJ, Monteiro R. Metabolically healthy obesity-heterogeneity in definitions and unconventional factors. Metabolites. 2020;10(2):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Neeland IJ, Poirier P, Després JP. Cardiovascular and metabolic heterogeneity of obesity: clinical challenges and implications for management. Circulation. 2018;137(13):1391–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sulc J, Winkler TW, Heid IM, Kutalik Z. Heterogeneity in obesity: genetic basis and metabolic consequences. Curr Diab Rep. 2020;20(1):1. [DOI] [PubMed] [Google Scholar]
  • 10.Valencak TG, Osterrieder A, Schulz TJ. Sex matters: the effects of biological sex on adipose tissue biology and energy metabolism. Redox Biol. 2017;12:806–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Peters SAE, Bots SH, Woodward M. Sex differences in the association between measures of general and central adiposity and the risk of myocardial infarction: results from the UK Biobank. J Am Heart Assoc. 2018;7(5):e008507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Caceres BA, Streed CG Jr, Corliss HL, Lloyd-Jones DM, Matthews PA, Mukherjee M, Poteat T, Rosendale N, Ross LM. Assessing and addressing cardiovascular health in LGBTQ adults: a scientific statement from the American Heart Association. Circulation. 2020;142(19):e321–e332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Palaniappan LP, Araneta MR, Assimes TL, Barrett-Connor EL, Carnethon MR, Criqui MH, Fung GL, Narayan KM, Patel H, Taylor-Piliae REet al. . Call to action: cardiovascular disease in Asian Americans: a science advisory from the American Heart Association. Circulation. 2010;122(12):1242–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rodriguez CJ, Allison M, Daviglus ML, Isasi CR, Keller C, Leira EC, Palaniappan L, Piña IL, Ramirez SM, Rodriguez Bet al. . Status of cardiovascular disease and stroke in Hispanics/Latinos in the United States: a science advisory from the American Heart Association. Circulation. 2014;130(7):593–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Carnethon MR, Pu J, Howard G, Albert MA, Anderson CAM, Bertoni AG, Mujahid MS, Palaniappan L, Taylor HA Jr, Willis Met al. . Cardiovascular health in African Americans: a scientific statement from the American Heart Association. Circulation. 2017;136(21):e393–423. [DOI] [PubMed] [Google Scholar]
  • 16.Churchwell K, Elkind MSV, Benjamin RM, Carson AP, Chang EK, Lawrence W, Mills A, Odom TM, Rodriguez CJ, Rodriguez Fet al. . Call to Action: Structural Racism as a Fundamental Driver of Health Disparities: A Presidential Advisory From the American Heart Association. Circulation. 2020;142(24):e454–e468. [DOI] [PubMed] [Google Scholar]
  • 17.Chung ST, Courville AB, Onuzuruike AU, Galvan-De La Cruz M, Mabundo LS, DuBose CW, Kasturi K, Cai H, Gharib AM, Walter PJet al. . Gluconeogenesis and risk for fasting hyperglycemia in Black and White women. JCI Insight. 2018;3(18):1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Luo J, Hodge A, Hendryx M, Byles JE. Age of obesity onset, cumulative obesity exposure over early adulthood and risk of type 2 diabetes. Diabetologia. 2020;63(3):519–27. [DOI] [PubMed] [Google Scholar]
  • 19.Malhotra S, Sivasubramanian R, Singhal V. Adult obesity and its complications: a pediatric disease?. Curr Opin Endocrinol, Diabetes Obes. 2021;28(1):46–54. [DOI] [PubMed] [Google Scholar]
  • 20.Dutta S, Sengupta P. Men and mice: relating their ages. Life Sci. 2016;152:244–8. [DOI] [PubMed] [Google Scholar]
  • 21.Zeisel SH. Diet-gene interactions underlie metabolic individuality and influence brain development: implications for clinical practice derived from studies on choline metabolism. Ann Nutr Metab. 2012;60(Suppl 3):19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wildman RP, Muntner P, Reynolds K, McGinn AP, Rajpathak S, Wylie-Rosett J, Sowers MR. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999–2004). Arch Intern Med. 2008;168(15):1617–24. [DOI] [PubMed] [Google Scholar]
  • 23.Shin MJ, Hyun YJ, Kim OY, Kim JY, Jang Y, Lee JH.. Weight loss effect on inflammation and LDL oxidation in metabolically healthy but obese (MHO) individuals: low inflammation and LDL oxidation in MHO women. Int J Obes. 2006;30(10):1529–34. [DOI] [PubMed] [Google Scholar]
  • 24.Johnson AR, Milner JJ, Makowski L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunol Rev. 2012;249(1):218–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lackey DE, Olefsky JM. Regulation of metabolism by the innate immune system. Nat Rev Endocrinol. 2016;12(1):15–28. [DOI] [PubMed] [Google Scholar]
  • 26.Reaven GM. Why Syndrome X? From Harold Himsworth to the insulin resistance syndrome. Cell Metab. 2005;1(1):9–14. [DOI] [PubMed] [Google Scholar]
  • 27.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LAet al. . Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ujvari B, Jacqueline C, Misse D, Amar V, Fitzpatrick JC, Jennings G, Beckmann C, Rome S, Biro PA, Gatenby Ret al. . Obesity paradox in cancer: is bigger really better? Evol Appl. 2019;12(6):1092–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Braun N, Hoess C, Kutz A, Christ-Crain M, Thomann R, Henzen C, Zimmerli W, Mueller B, Schuetz P. Obesity paradox in patients with community-acquired pneumonia: is inflammation the missing link?. Nutrition. 2017;33:304–10. [DOI] [PubMed] [Google Scholar]
  • 31.Rey-López JP, de Rezende LF, Pastor-Valero M, Tess BH.. The prevalence of metabolically healthy obesity: a systematic review and critical evaluation of the definitions used. Obes Rev. 2014;15(10):781–90. [DOI] [PubMed] [Google Scholar]
  • 32.Phillips CM. Metabolically healthy obesity: personalised and public health implications. Trends Endocrinol Metab. 2016;27(4):189–91. [DOI] [PubMed] [Google Scholar]
  • 33.Opio J, Croker E, Odongo GS, Attia J, Wynne K, McEvoy M. Metabolically healthy overweight/obesity are associated with increased risk of cardiovascular disease in adults, even in the absence of metabolic risk factors: a systematic review and meta-analysis of prospective cohort studies. Obes Rev. 2020;21(12):e13127. [DOI] [PubMed] [Google Scholar]
  • 34.Gao M, Lv J, Yu C, Guo Y, Bian Z, Yang R, Du H, Yang L, Chen Y, Li Zet al. . Metabolically healthy obesity, transition to unhealthy metabolic status, and vascular disease in Chinese adults: a cohort study. PLoS Med. 2020;17(10):e1003351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: facts and fantasies. J Clin Invest. 2019;129(10):3978–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Newgard CB. Metabolomics and metabolic diseases: where do we stand?. Cell Metab. 2017;25(1):43–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.de Toro-Martin J, Arsenault BJ, Despres JP, Vohl MC. Precision nutrition: a review of personalized nutritional approaches for the prevention and management of metabolic syndrome. Nutrients. 2017;9(8):913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Carnethon MR, Sternfeld B, Schreiner PJ, Jacobs DR Jr., Lewis CE, Liu K, Sidney S. Association of 20-year changes in cardiorespiratory fitness with incident type 2 diabetes: the coronary artery risk development in young adults (CARDIA) fitness study. Diabetes Care. 2009;32(7):1284–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chow LS, Odegaard AO, Bosch TA, Bantle AE, Wang Q, Hughes J, Carnethon M, Ingram KH, Durant N, Lewis CEet al. . Twenty year fitness trends in young adults and incidence of prediabetes and diabetes: the CARDIA study. Diabetologia. 2016;59(8):1659–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Murthy VL, Abbasi SA, Siddique J, Colangelo LA, Reis J, Venkatesh BA, Carr JJ, Terry JG, Camhi SM, Jerosch-Herold Met al. . Transitions in metabolic risk and long-term cardiovascular health: Coronary Artery Risk Development in Young Adults (CARDIA) study. J Am Heart Assoc. 2016;5(10):934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Reis JP, Allen N, Gunderson EP, Lee JM, Lewis CE, Loria CM, Powell-Wiley TM, Rana JS, Sidney S, Wei Get al. . Excess body mass index- and waist circumference-years and incident cardiovascular disease: the CARDIA study. Obesity. 2015;23(4):879–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Reis JP, Hankinson AL, Loria CM, Lewis CE, Powell-Wiley T, Wei GS, Liu K. Duration of abdominal obesity beginning in young adulthood and incident diabetes through middle age: the CARDIA study. Diabetes Care. 2013;36(5):1241–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Poirier P, Giles TD, Bray GA, Hong Y, Stern JS, Pi-Sunyer FX, Eckel RH. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation. 2006;113(6):898–918. [DOI] [PubMed] [Google Scholar]
  • 44.Scherer PE, Hill JA. Obesity, diabetes, and cardiovascular diseases: a compendium. Circ Res. 2016;118(11):1703–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hunter WG, Kelly JP, McGarrah RW 3rd, Kraus WE, Shah SH.. Metabolic dysfunction in heart failure: diagnostic, prognostic, and pathophysiologic insights from metabolomic profiling. Curr Heart Fail Rep. 2016;13(3):119–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shah SH, Newgard CB. Integrated metabolomics and genomics: systems approaches to biomarkers and mechanisms of cardiovascular disease. Circ Cardiovasc Genet. 2015;8(2):410–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Shah SH, Kraus WE, Newgard CB. Metabolomic profiling for the identification of novel biomarkers and mechanisms related to common cardiovascular diseases: form and function. Circulation. 2012;126(9):1110–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.McGarrah RW, Crown SB, Zhang GF, Shah SH, Newgard CB. Cardiovascular metabolomics. Circ Res. 2018;122(9):1238–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Musunuru K, Ingelsson E, Fornage M, Liu P, Murphy AM, Newby LK, Newton-Cheh C, Perez MV, Voora D, Woo D. The expressed genome in cardiovascular diseases and stroke: refinement, diagnosis, and prediction: a scientific statement from the American Heart Association. Circ Cardiovasc Genet. 2017;10(4):e000037. [DOI] [PubMed] [Google Scholar]
  • 50.Cheng Z, Zheng L, Almeida FA. Epigenetic reprogramming in metabolic disorders: nutritional factors and beyond. J Nutr Biochem. 2018;54:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 2019;29(5):1028–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ouni M, Schürmann A.. Epigenetic contribution to obesity. Mamm Genome. 2020;31(5–6):134–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kwok KH, Lam KS, Xu A. Heterogeneity of white adipose tissue: molecular basis and clinical implications. Exp Mol Med. 2016;48:e215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nic-Can GI, BA Rodas-Junco, Carrillo-Cocom LM, Zepeda-Pedreguera A, Penaloza-Cuevas R, Aguilar-Ayala FJ, Rojas-Herrera RA. Epigenetic regulation of adipogenic differentiation by histone lysine demethylation. Int J Mol Sci. 2019;20(16):3918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Aspects Med. 2012;33(1):26–34. [DOI] [PubMed] [Google Scholar]
  • 56.Ravussin E, Galgani JE. The implication of brown adipose tissue for humans. Annu Rev Nutr. 2011;31:33–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kalupahana NS, Goonapienuwala BL, Moustaid-Moussa N. Omega-3 fatty acids and adipose tissue: inflammation and browning. Annu Rev Nutr. 2020;40:25–49. [DOI] [PubMed] [Google Scholar]
  • 58.Fried SK, Lee MJ, Karastergiou K. Shaping fat distribution: new insights into the molecular determinants of depot- and sex-dependent adipose biology. Obesity. 2015;23(7):1345–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Unger RH, Scherer PE. Gluttony, sloth and the metabolic syndrome: a roadmap to lipotoxicity. Trends Endocrinol Metab. 2010;21(6):345–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rutkowski JM, Stern JH, Scherer PE. The cell biology of fat expansion. J Cell Biol. 2015;208(5):501–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cleal L, Aldea T, Chau YY. Fifty shades of white: understanding heterogeneity in white adipose stem cells. Adipocyte. 2017;6(3):205–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Blüher M, Wilson-Fritch L, Leszyk J, Laustsen PG, Corvera S, Kahn CR. Role of insulin action and cell size on protein expression patterns in adipocytes. J Biol Chem. 2004;279(30):31902–9. [DOI] [PubMed] [Google Scholar]
  • 63.Yvan-Charvet L, Massiéra F, Lamandé N, Ailhaud G, Teboul M, Moustaid-Moussa N, Gasc JM, Quignard-Boulangé A. Deficiency of angiotensin type 2 receptor rescues obesity but not hypertension induced by overexpression of angiotensinogen in adipose tissue. Endocrinology. 2009;150(3):1421–8. [DOI] [PubMed] [Google Scholar]
  • 64.Weinstock A, Moura Silva H, Moore KJ, Schmidt AM, Fisher EA. Leukocyte heterogeneity in adipose tissue, including in obesity. Circ Res. 2020;126(11):1590–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, Powell C, Vedantam S, Buchkovich ML, Yang Jet al. . Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shungin D, Winkler TW, Croteau-Chonka DC, Ferreira T, Locke AE, Magi R, Strawbridge RJ, Pers TH, Fischer K, Justice AEet al. . New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518(7538):187–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Monda KL, Chen GK, Taylor KC, Palmer C, Edwards TL, Lange LA, Ng MC, Adeyemo AA, Allison MA, Bielak LFet al. . A meta-analysis identifies new loci associated with body mass index in individuals of African ancestry. Nat Genet. 2013;45(6):690–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, Lango Allen H, Lindgren CM, Luan J, Magi Ret al. . Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet. 2010;42(11):937–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, Thorleifsson G, Zillikens MC, Speliotes EK, Magi Ret al. . Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nat Genet. 2010;42(11):949–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, Berndt SI, Elliott AL, Jackson AU, Lamina Cet al. . Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat Genet. 2009;41(1):25–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Loos RJ, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP, Beckmann JSet al. . Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet. 2008;40(6):768–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lindgren CM, Heid IM, Randall JC, Lamina C, Steinthorsdottir V, Qi L, Speliotes EK, Thorleifsson G, Willer CJ, Herrera BMet al. . Genome-wide association scan meta-analysis identifies three loci influencing adiposity and fat distribution. PLos Genet. 2009;5(6):e1000508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Liu CT, Monda KL, Taylor KC, Lange L, Demerath EW, Palmas W, Wojczynski MK, Ellis JC, Vitolins MZ, Liu Set al. . Genome-wide association of body fat distribution in African ancestry populations suggests new loci. PLos Genet. 2013;9(8):e1003681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kilpelainen TO, Zillikens MC, Stancakova A, Finucane FM, Ried JS, Langenberg C, Zhang W, Beckmann JS, Luan J, Vandenput Let al. . Genetic variation near IRS1 associates with reduced adiposity and an impaired metabolic profile. Nat Genet. 2011;43(8):753–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Yang J, Loos RJ, Powell JE, Medland SE, Speliotes EK, Chasman DI, Rose LM, Thorleifsson G, Steinthorsdottir V, Magi Ret al. . FTO genotype is associated with phenotypic variability of body mass index. Nature. 2012;490(7419):267–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Carlson CS, Matise TC, North KE, Haiman CA, Fesinmeyer MD, Buyske S, Schumacher FR, Peters U, Franceschini N, Ritchie MDet al. . Generalization and dilution of association results from European GWAS in populations of non-European ancestry: the PAGE study. PLoS Biol. 2013;11(9):e1001661. Epub 2013 Sep 17. doi:10.1371/journal.pbio.1001661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wojcik GL, Graff M, Nishimura KK, Tao R, Haessler J, Gignoux CR, Highland HM, Patel YM, Sorokin EP, Avery CLet al. . Genetic analyses of diverse populations improves discovery for complex traits. Nature. 2019;570(7762):514–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Matise TC, Ambite JL, Buyske S, Carlson CS, Cole SA, Crawford DC, Haiman CA, Heiss G, Kooperberg C, Marchand LLet al. . The next PAGE in understanding complex traits: design for the analysis of Population Architecture Using Genetics and Epidemiology (PAGE) study. Am J Epidemiol. 2011;174(7):849–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Martin AR, Gignoux CR, Walters RK, Wojcik GL, Neale BM, Gravel S, Daly MJ, Bustamante CD, Kenny EE. Human demographic history impacts genetic risk prediction across diverse populations. Am J Hum Genet. 2017;100(4):635–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Martin AR, Kanai M, Kamatani Y, Okada Y, Neale BM, Daly MJ. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat Genet. 2019;51(4):584–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Hoffmann TJ, Choquet H, Yin J, Banda Y, Kvale MN, Glymour M, Schaefer C, Risch N, Jorgenson E. A large multiethnic genome-wide association study of adult body mass index identifies novel loci. Genetics. 2018;210(2):499–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.GIANT. GIANT consortium data files 2019 [02/16/2021] [Internet]. Available from: https://portals.broadinstitute.org/collaboration/giant/index.php?title=GIANT_consortium_data_files&oldid=579.
  • 83.Saul MC, Philip VM, Reinholdt LG, Chesler EJ. High-diversity mouse populations for complex traits. Trends Genet. 2019;35(7):501–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Perlman RL. Mouse models of human disease: an evolutionary perspective. Evol Med Public Health. 2016;2016(1):170–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Attie AD, Churchill GA, Nadeau JH.. How mice are indispensable for understanding obesity and diabetes genetics. Curr Opin Endocrinol Diabetes Obes. 2017;24(2):83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Churchill GA, Gatti DM, Munger SC, Svenson KL. The diversity outbred mouse population. Mamm Genome. 2012;23(9–10):713–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.French JE, Gatti DM, Morgan DL, Kissling GE, Shockley KR, Knudsen GA, Shepard KG, Price HC, King D, Witt KLet al. . Diversity outbred mice identify population-based exposure thresholds and genetic factors that influence benzene-induced genotoxicity. Environ Health Perspect. 2015;123(3):237–45. Epub 2014 Nov 6. doi:10.1289/ehp.1408202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lusis AJ, Seldin MM, Allayee H, Bennett BJ, Civelek M, Davis RC, Eskin E, Farber CR, Hui S, Mehrabian Met al. . The Hybrid Mouse Diversity Panel: a resource for systems genetics analyses of metabolic and cardiovascular traits. J Lipid Res. 2016;57(6):925–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Smallwood TL, Gatti DM, Quizon P, Weinstock GM, Jung KC, Zhao L, Hua K, Pomp D, Bennett BJ.. High-resolution genetic mapping in the diversity outbred mouse population identifies Apobec1 as a candidate gene for atherosclerosis. G3. 2014;4(12):2353–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Keller MP, Gatti DM, Schueler KL, Rabaglia ME, Stapleton DS, Simecek P, Vincent M, Allen S, Broman AT, Bacher Ret al. . Genetic drivers of pancreatic islet function. Genetics. 2018;209(1):335–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Frazer KA, Eskin E, Kang HM, Bogue MA, Hinds DA, Beilharz EJ, Gupta RV, Montgomery J, Morenzoni MM, Nilsen GBet al. . A sequence-based variation map of 8.27 million SNPs in inbred mouse strains. Nature. 2007;448(7157):1050–3. [DOI] [PubMed] [Google Scholar]
  • 92.Yang H, Bell TA, Churchill GA, Pardo-Manuel de Villena F. On the subspecific origin of the laboratory mouse. Nat Genet. 2007;39(9):1100–7. [DOI] [PubMed] [Google Scholar]
  • 93.Collaborative Cross Consortium . The genome architecture of the Collaborative Cross mouse genetic reference population. Genetics. 2012;190(2):389–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Svenson KL, Gatti DM, Valdar W, Welsh CE, Cheng R, Chesler EJ, Palmer AA, McMillan L, Churchill GA. High-resolution genetic mapping using the mouse diversity outbred population. Genetics. 2012;190(2):437–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Broman KW, Gatti DM, Svenson KL, Sen Ś, Churchill GA.. Cleaning genotype data from diversity outbred mice. G3 (Bethesda). 2019;9(5):1571–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Corty RW, Kumar V, Tarantino LM, Takahashi JS, Valdar W. Mean-variance QTL mapping identifies novel QTL for circadian activity and exploratory behavior in mice. G3 (Bethesda). 2018;8(12):3783–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Bogue MA, Churchill GA, Chesler EJ. Collaborative cross and diversity outbred data resources in the mouse phenome database. Mamm Genome. 2015;26(9–10):511–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Bult CJ, Blake JA, Smith CL, Kadin JA, Richardson JE. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 2019;47(D1):D801–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pal A, Al-Shaer AE, Guesdon W, Torres MJ, Armstrong M, Quinn K, Davis T, Reisdorph N, Neufer PD, Spangenburg EEet al. . Resolvin E1 derived from eicosapentaenoic acid prevents hyperinsulinemia and hyperglycemia in a host genetic manner. FASEB J. 2020;34(8):10640–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Aylor DL, Valdar W, Foulds-Mathes W, Buus RJ, Verdugo RA, Baric RS, Ferris MT, Frelinger JA, Heise M, Frieman MBet al. . Genetic analysis of complex traits in the emerging collaborative cross. Genome Res. 2011;21(8):1213–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kelada SN, Carpenter DE, Aylor DL, Chines P, Rutledge H, Chesler EJ, Churchill GA, Pardo-Manuel de Villena F, Schwartz DA, Collins FS. Integrative genetic analysis of allergic inflammation in the murine lung. Am J Respir Cell Mol Biol. 2014;51(3):436–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Phillippi J, Xie Y, Miller DR, Bell TA, Zhang Z, Lenarcic AB, Aylor DL, Krovi SH, Threadgill DW, de Villena FPet al. . Using the emerging collaborative cross to probe the immune system. Genes Immun. 2014;15(1):38–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Himes BE, Sheppard K, Berndt A, Leme AS, Myers RA, Gignoux CR, Levin AM, Gauderman WJ, Yang JJ, Mathias RAet al. . Integration of mouse and human genome-wide association data identifies KCNIP4 as an asthma gene. PLoS One. 2013;8(2):e56179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang Y, Wang H, Howard AG, Tsilimigras MCB, Avery CL, Meyer KA, Sha W, Sun S, Zhang J, Su Cet al. . Gut microbiota and host plasma metabolites in association with blood pressure in Chinese adults. Hypertension. 2021;77(2):706–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mungall CJ, McMurry JA, Köhler S, Balhoff JP, Borromeo C, Brush M, Carbon S, Conlin T, Dunn N, Engelstad Met al. . The Monarch Initiative: an integrative data and analytic platform connecting phenotypes to genotypes across species. Nucleic Acids Res. 2017;45(D1):D712–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ahlqvist E, Storm P, Käräjämäki A, Martinell M, Dorkhan M, Carlsson A, Vikman P, Prasad RB, Aly DM, Almgren Pet al. . Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6(5):361–9. [DOI] [PubMed] [Google Scholar]
  • 107.Ahlqvist E, Prasad RB, Groop L. Subtypes of type 2 diabetes determined from clinical parameters. Diabetes. 2020;69(10):2086–93. [DOI] [PubMed] [Google Scholar]
  • 108.Udler MS, Kim J, von Grotthuss M, Bonàs-Guarch S, Cole JB, Chiou J, Boehnke M, Laakso M, Atzmon G, Glaser Bet al. . Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: a soft clustering analysis. PLoS Med. 2018;15(9):e1002654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hunter DJ. Gene-environment interactions in human diseases. Nat Rev Genet. 2005;6(4):287–98. [DOI] [PubMed] [Google Scholar]
  • 110.Davey Smith G. Use of genetic markers and gene-diet interactions for interrogating population-level causal influences of diet on health. Genes Nutr. 2011;6(1):27–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mathers JC. Nutrigenomics in the modern era. Proc Nutr Soc. 2017;76(3):265–75. [DOI] [PubMed] [Google Scholar]
  • 112.Marti A, Martinez-González MA, Martinez JA.. Interaction between genes and lifestyle factors on obesity. Proc Nutr Soc. 2008;67(1):1–8. [DOI] [PubMed] [Google Scholar]
  • 113.Reddon H, Guéant JL, Meyre D. The importance of gene-environment interactions in human obesity. Clin Sci. 2016;130(18):1571–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.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. 2017;13(9):e1006977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Castillo JJ, Orlando RA, Garver WS. Gene-nutrient interactions and susceptibility to human obesity. Genes Nutr. 2017;12:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Heianza Y, Qi L. Gene-diet interaction and precision nutrition in obesity. Int J Mol Sci. 2017;18(4):787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ordovas JM, Tai ES. Why study gene-environment interactions?. Curr Opin Lipidol. 2008;19(2):158–67. [DOI] [PubMed] [Google Scholar]
  • 118.Smith CE, Tucker KL, Arnett DK, Noel SE, Corella D, Borecki IB, Feitosa MF, Aslibekyan S, Parnell LD, Lai CQet al. . Apolipoprotein A2 polymorphism interacts with intakes of dairy foods to influence body weight in 2 U.S. populations. J Nutr. 2013;143(12):1865–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Czajkowski P, Adamska-Patruno E, Bauer W, Fiedorczuk J, Krasowska U, Moroz M, Gorska M, Kretowski A. The impact of FTO genetic variants on obesity and its metabolic consequences is dependent on daily macronutrient intake. Nutrients. 2020;12(11):3255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Koochakpoor G, Hosseini-Esfahani F, Daneshpour MS, Hosseini SA, Mirmiran P. Effect of interactions of polymorphisms in the Melanocortin-4 receptor gene with dietary factors on the risk of obesity and type 2 diabetes: a systematic review. Diabet Med. 2016;33(8):1026–34. [DOI] [PubMed] [Google Scholar]
  • 121.Brahe LK, Ängquist L, Larsen LH, Vimaleswaran KS, Hager J, Viguerie N, Loos RJ, Handjieva-Darlenska T, Jebb SA, Hlavaty Pet al. . Influence of SNPs in nutrient-sensitive candidate genes and gene-diet interactions on blood lipids: the DiOGenes study. Br J Nutr. 2013;110(5):790–6. [DOI] [PubMed] [Google Scholar]
  • 122.Cecil JE, Palmer CN, Fischer B, Watt P, Wallis DJ, Murrie I, Hetherington MM. Variants of the peroxisome proliferator-activated receptor gamma- and beta-adrenergic receptor genes are associated with measures of compensatory eating behaviors in young children. Am J Clin Nutr. 2007;86(1):167–73. [DOI] [PubMed] [Google Scholar]
  • 123.Qi L. Gene-diet interactions in complex disease: current findings and relevance for public health. Curr Nutr Rep. 2012;1(4):222–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Price AL, Spencer CC, Donnelly P. Progress and promise in understanding the genetic basis of common diseases. Proc Biol Sci. 2015;282(1821):20151684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Graff M, Scott RA, Justice AE, Young KL, Feitosa MF, Barata L, Winkler TW, Chu AY, Mahajan A, Hadley Det al. . Genome-wide physical activity interactions in adiposity—a meta-analysis of 200,452 adults. PLos Genet. 2017;13(4):e1006528. Epub 2017 Apr 27. doi:10.1371/journal.pgen.1006528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ahmad S, Rukh G, Varga TV, Ali A, Kurbasic A, Shungin D, Ericson U, Koivula RW, Chu AY, Rose LMet al. . Gene × physical activity interactions in obesity: combined analysis of 111,421 individuals of European ancestry. PLos Genet. 2013;9(7):e1003607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Reddon H, Gerstein HC, Engert JC, Mohan V, Bosch J, Desai D, Bailey SD, Diaz R, Yusuf S, Anand SSet al. . Physical activity and genetic predisposition to obesity in a multiethnic longitudinal study. Sci Rep. 2016;6:18672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Moon JY, Wang T, Sofer T, North KE, Isasi CR, Cai J, Gellman MD, Moncrieft AE, Sotres-Alvarez D, Argos Met al. . Objectively measured physical activity, sedentary behavior, and genetic predisposition to obesity in U.S. Hispanics/Latinos: results from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Diabetes. 2017;66(12):3001–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Celis-Morales C, Lyall DM, Guo Y, Steell L, Llanas D, Ward J, Mackay DF, Biello SM, Bailey ME, Pell JPet al. . Sleep characteristics modify the association of genetic predisposition with obesity and anthropometric measurements in 119,679 UK Biobank participants. Am J Clin Nutr. 2017;105(4):980–90. [DOI] [PubMed] [Google Scholar]
  • 130.Watson NF, Harden KP, Buchwald D, Vitiello MV, Pack AI, Weigle DS, Goldberg J.. Sleep duration and body mass index in twins: a gene-environment interaction. Sleep. 2012;35(5):597–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sun Y, Fang J, Wan Y, Hu J, Xu Y, Tao F.. Polygenic differential susceptibility to cumulative stress exposure and childhood obesity. Int J Obes. 2018;42(6):1177–84. [DOI] [PubMed] [Google Scholar]
  • 132.Fesinmeyer MD, North KE, Lim U, Bůžková P, Crawford DC, Haessler J, Gross MD, Fowke JH, Goodloe R, Love SAet al. . Effects of smoking on the genetic risk of obesity: the population architecture using genomics and epidemiology study. BMC Med Genet. 2013;14:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Martin LJ, Kissebah AH, Sonnenberg GE, Blangero J, Comuzzie AG. Genotype-by-smoking interaction for leptin levels in the Metabolic Risk Complications of Obesity Genes project. Int J Obes. 2003;27(3):334–40. [DOI] [PubMed] [Google Scholar]
  • 134.Rohde JF, Ängquist L, Larsen SC, Tolstrup JS, Husemoen LLN, Linneberg A, Toft U, Overvad K, Halkjær J, Tjønneland Aet al. . Alcohol consumption and its interaction with adiposity-associated genetic variants in relation to subsequent changes in waist circumference and body weight. Nutr J. 2017;16(1):51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Foraita R, Günther F, Gwozdz W, Reisch LA, Russo P, Lauria F, Siani A, Veidebaum T, Tornaritis M, Iacoviello Let al. . Does the FTO gene interact with the socioeconomic status on the obesity development among young European children? Results from the IDEFICS study. Int J Obes. 2015;39(1):1–6. [DOI] [PubMed] [Google Scholar]
  • 136.Midha KK, Chakraborty BS, Ganes DA, Hawes EM, Hubbard JW, Keegan DL, Korchinski ED, McKay G.. Intersubject variation in the pharmacokinetics of haloperidol and reduced haloperidol. J Clin Psychopharmacol. 1989;9(2):98–104. [DOI] [PubMed] [Google Scholar]
  • 137.Pereira P, Bandeira A, Kleine B, Marquez C, Coutinho A, Martinez C.. A model system for the analysis of B-cell activation and effector T-cell functions. T cell-dependent B-cell responses facilitated by anti-I-A antibodies. Scand J Immunol. 1989;29(1):49–56. [DOI] [PubMed] [Google Scholar]
  • 138.van Vliet-Ostaptchouk JV, Snieder H, Lagou V.. Gene-lifestyle interactions in obesity. Curr Nutr Rep. 2012;1(3):184–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Eldridge AL, Piernas C, Illner AK, Gibney MJ, Gurinović MA, de Vries JHM, Cade JE.. Evaluation of new technology-based tools for dietary intake assessment—an ILSI Europe Dietary Intake and Exposure Task Force evaluation. Nutrients. 2018;11(1):55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bell BM, Alam R, Alshurafa N, Thomaz E, Mondol AS, de la Haye K, Stankovic JA, Lach J, Spruijt-Metz D.. Automatic, wearable-based, in-field eating detection approaches for public health research: a scoping review. NPJ Digit Med. 2020;3:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Khazen W, Jeanne JF, Demaretz L, Schäfer F, Fagherazzi G.. Rethinking the use of mobile apps for dietary assessment in medical research. J Med Internet Res. 2020;22(6):e15619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Warden CH, Fisler JS.. Comparisons of diets used in animal models of high-fat feeding. Cell Metab. 2008;7(4):277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Hintze KJ, Benninghoff AD, Cho CE, Ward RE.. Modeling the Western diet for preclinical investigations. Adv Nutr. 2018;9(3):263–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Landis SC, Amara SG, Asadullah K, Austin CP, Blumenstein R, Bradley EW, Crystal RG, Darnell RB, Ferrante RJ, Fillit Het al. . A call for transparent reporting to optimize the predictive value of preclinical research. Nature. 2012;490(7419):187–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, Browne WJ, Clark A, Cuthill IC, Dirnagl Uet al. . The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. PLoS Biol. 2020;18(7):e3000410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Speakman JR. Use of high-fat diets to study rodent obesity as a model of human obesity. Int J Obes. 2019;43(8):1491–2. [DOI] [PubMed] [Google Scholar]

Articles from Advances in Nutrition are provided here courtesy of American Society for Nutrition

RESOURCES