Prev Chronic DisPreventing Chronic Disease1545-1151Centers for Disease Control and Prevention195275972722400PCDv63_09_0011Special TopicPeer ReviewedThe Effect of Neurohormonal Factors, Epigenetic Factors, and Gut Microbiota on Risk of ObesityHaemerMatthew A.MDUniversity of Colorado School of Medicine, Pediatrics Section of Nutrition, The Children’s Hospital
Haemer.matthew@tchden.org13123 E 16th Ave, B270, Aurora, CO 80045720-777-7474
HuangTerry T.PhD, MPHObesity Research Strategic Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MarylandDanielsStephen R.MD, PhDDaniels, University of Colorado School of Medicine and The Children’s Hospital, Denver, Colorado
72009156200963A96

Molecular, cellular, and epidemiologic findings suggest that neurohormonal, epigenetic, and microbiologic mechanisms may influence risk for obesity by interacting with socioenvironmental factors. Homeostatic and nonhomeostatic neural controls of energy predispose people to obesity, and this predisposition may be exaggerated by the influence of media, marketing, and sleep patterns. Epigenetic gene regulation may account for the influence of modifiable early life or maternal exposures on obesity risk. Alterations in gut flora caused by infant feeding practices or later diet may influence the absorption and storage of energy. Further exploration of how these molecular-cellular mechanisms might increase obesity risk in response to modifiable socioeconomic factors requires the partnership of laboratory and public health researchers.

Introduction

We describe components of a novel paradigm for obesity-related research and public health interventions that are designed to generate cross-disciplinary hypotheses that account for multiple levels of causation from molecular to societal. The goals of this approach are to understand the causal pathways leading to patterns of obesity in populations and to identify where interventions that can broadly affect the population can be implemented (1). Our focus is on the mechanisms that may be useful for understanding how socioenvironmental factors interact with biological processes to affect energy balance. We review neurohormonal controls, epigenetics, and microbiologic mechanisms in gut flora that may influence risk for obesity.

Neurohormonal ControlsHomeostatic controls

The complex neurohormonal systems controlling weight and adiposity can be categorized as either homeostatic or nonhomeostatic (2). Parsimonious homeostatic mechanisms provided a survival advantage to people faced with periodic starvation. Organisms that could consume, store, and conserve energy efficiently were likely to survive and reproduce (3). Although evolution may have selected against extreme adiposity (4), the negative feedback signals against excess intake are insufficient to maintain normal body weight for most humans who have easy access to palatable, calorie-dense foods (5).

When provided a diet high in calories, animals prone to obesity rapidly increase fat stores (2). Furthermore, when obese rats lose weight or have their calorie intake restricted, they mount the same neurohormonal drive as do lean rats to increase intake and decrease energy expenditure, effectively defending obesity (6,7). After weight loss, the average resting energy expenditure of obese people is markedly and persistently reduced (8). These factors are blamed for the weight regain that occurs in approximately 80% to 90% of obese people who have lost weight (8,9).

A complex interplay of neurotransmitters, hormones, and metabolites regulates food intake in the brain. Metabolic sensing neurons of the hypothalamus and other brain areas respond to signals of energy intake, demand, or storage including circulating glucose, leptin from adipocytes, insulin, ghrelin from the stomach, adrenal steroids, polypeptide YY from the intestines, fatty acids, ketones, lactate, vagal nerve afferents, and intrinsic neurotransmitters (5). Hypothalamic neurons release neurotransmitters that activate either catabolic processes (eg, a-melanocyte-stimulating hormone [a-MSH], cocaine- and amphetamine-regulated transcript [CART], corticotropin-releasing hormone [CRH]) or anabolic processes (eg, neuropeptide Y [NPY], agouti-related protein [AGRP], orexins) (10). Hypothalamic neurons signal broadly to the pituitary, brainstem, midbrain, and forebrain. These pathways regulating energy intake and expenditure have been demonstrated experimentally in animal models and through functional neuroimaging in humans (11).

The interplay between the sympathetic nervous system and leptin signaling is an example of neurohormonal protection of adiposity. With calorie restriction, sympathetic activation releases glucose from glycogen stores and fatty acids from adipose tissue. Adipose tissue responds to sympathetic activity by markedly decreasing leptin production, which decreases resting energy expenditure and increases appetite to replenish fat stores (2). Some rare human obesity syndromes are associated with single gene defects within homeostatic pathways, including leptin deficiency, leptin receptor defect, pro-opiomelanocortin (POMC) deficit leading to impaired a-MSH production, and a-MSH receptor defects (12).

Nonhomeostatic controls

Nonhomeostatic systems promote excess weight gain through responses to the reward properties of food and psychosocial factors associated with eating (5). When a palatable and calorically dense diet is provided, rats eat far beyond limits of homeostasis and develop extreme levels of obesity, even rats predisposed to leanness (13). The more palatable the diet, the higher the degree of obesity and the longer it is sustained (13).

The organization and function of the human brain is a reflection that throughout all but the most recent evolutionary history, obtaining food was a difficult task. Complex pathways record past context and the expectation of reward (14). Reward properties of foods (the stimuli that augment the drive to obtain foods) mediate "liking" by action at opioid receptors and "wanting" by action at dopaminergic receptors. Both of these receptors also mediate addiction (4). Metabolic signals modify the sensing thresholds for food-related stimuli, food-seeking behavior, and reward signals (4). Chronic stress enhances the reward value of foods (15). Although the subcortical areas contribute to subconscious drives for intake, the cortical areas integrate these underlying signals with learned motivational cues. These cues can drive intake well beyond subcortical demands of energy needs (16). Overall, the drive to eat is the result of complex, redundant systems that protect against starvation, but the systems are grossly mismatched to the food and activity environment of the developed world (17).

Implications of neurohormonal mechanisms for obesity and public health

The potency of homeostatic and nonhomeostatic forces that promote weight gain and prevent weight loss make clear the value of obesity prevention, especially considering the cost and difficulty of obesity treatment. Many socioenvironmental factors interact with the neurologic drivers of intake, so identifying and intervening on modifiable levels to prevent obesity is a challenging task.

Sleep may play a role in maintaining proper energy balance by influencing neurohormonal controls (18). A meta-analysis of 30 cross-sectional studies through early 2008 found an odds ratio for obesity of 1.89 in children with short duration of sleep and 1.55 in adults with short duration of sleep (19). More recently published longitudinal studies, one with 32 years of follow-up from birth, also found that shorter sleep times in childhood were significantly associated with increased body mass index (20,21). Experimental studies of sleep deprivation show increased hunger and appetite associated with neurohormonal mechanisms that promote intake: decreased levels of leptin, increased levels of ghrelin, increased sympathetic tone, and increased cortisol (22). Nonhormonal effects of short sleep, such as fatigue and decreased volitional energy expenditure, may also play a role in the association between sleep and obesity. Debate remains about the strength of the evidence that poor sleep causes obesity, and interventions to decrease obesity by increasing sleep have yet to be reported (23).

Media exposure and food marketing either overpower homeostatic negative feedback or strongly amplify nonhomeostatic drivers of intake. Distracting stimuli, such as television viewing while eating, strongly increase intake, possibly by interfering with neural signals of satiety (24). In a controlled experiment, viewing children's food advertisements caused children to eat much larger portions of snack foods compared to children who watched nonfood advertisements, and the effect was significantly larger on obese children than on normal-weight children (25). Marketing often seeks to influence the emotional responses to food (26) and succeeds in altering the perceived reward value of foods (27). Most food advertisements targeting preschool children involve fast-food restaurants or sweetened cereals. These advertisements associate the products with fun and happiness in an attempt to create long-term customers through positive emotional associations with the product (28). The food industry targets children at stages of development critical to establishing future eating habits (28).

Analysis by the National Bureau of Economic Research estimated that eliminating fast-food restaurant advertising to children would reduce the prevalence of obesity by 18% (29). Some governments have restricted television advertising of food products to children, and some advertisers have voluntarily restricted advertisements (30). However, the recent proliferation of other digital media sources, including cell phones, mobile music devices, broadband video, instant messaging, videogames, and virtual worlds, has created a "marketing ecosystem" for advertisement of food (31,32), and the influence of marketing on intake may become more pervasive in the future.

Epigenetics

Studies searching for determinants of risks for obesity and cardiovascular disease have found that genetics (33,34) and behavioral exposures (35) explain only part of the risk. Epigenetic mechanisms describe environment-gene interactions that may explain some residual risk. The term epigenetics refers to cellular mechanisms that affect gene expression without changing DNA sequence (36). Epigenetic markings can be inherited and modified throughout the lifespan (37). Epigenetic modifications during critical early periods, such as embryogenesis (38) have the most effect on phenotype. Fetal and early life exposures have been associated with numerous health outcomes later in life, including obesity (39,40). Changes to DNA marking and packaging may explain the influence of the environment on gene expression throughout a person's life and even across generations (41). Evidence is mounting from experimental studies in animals and from human epidemiologic studies that epigenetic mechanisms may affect risk for chronic disease, especially when the environment predicted by fetal experience does not match the environment later in life (42,43).

Animal models of epigenetics related to obesity

Experimental studies have subjected animals to dietary, chemical, and stress exposures during prenatal or early life that lead to epigenetic changes in gene expression in adulthood. The epigenetic changes caused by some exposures are preventable by other agents. Some changes in gene expression persist across generations when epigenetic markings are incompletely erased during formation of the sperm and ova (38). Several animal studies illustrate epigenetic influences on obesity.

The Agouti (Avy/a) mouse is a well-described model of epigenetically controlled obesity. Among mice carrying 1 obese allele (Avy) and 1 nonobese allele (a), those with inadequate methylation of the Avy allele develop obesity and yellow fur (38). The Avy protein blocks satiety signals from insulin and leptin on the hypothalamus (37). Obesity is amplified through multiple generations of Avy/a mice, but supplementing the diet with methyl donors prevents this amplification (44). Maternal ingestion of bisphenol A (BPA), a chemical used in polycarbonate plastic and epoxy resins, decreased methylation of the Avy allele in offspring (45). This decrease in methylation did not occur when the soy isoflavone genistein or a methyl donor, such as folic acid or vitamin B12, was added to the BPA-containing diet (45).

Rats whose mothers do not eat enough protein during pregnancy have decreased methylation and have increased expression of glucocorticoid receptors and peroxisomal proliferator-activated receptor-a in the liver. These changes are associated with components of metabolic syndrome including hypertension, dyslipidemia, and insulin resistance. Giving mothers folic acid supplements prevented the hypomethylation and normalized the gene expression in offspring (46).

Elevated reactivity to psychological stress has been associated with obesity in humans (47). In a model of early life stress, rats who are poorly nurtured by their mothers develop exaggerated stress responses and poorly nurture their own offspring; the pattern is repeated through multiple generations (48). Poor nurturing increases stress reactivity through methylation and de-acetylation of the glucocorticoid receptor gene in the brain, decreasing receptor production (49). Infusion of histone deacetylase inhibitors or "adopting out" offspring to highly nurturing mothers prevented these changes (48). Epigenetic recording of early parenting interactions affects the phenotype throughout the lifespan in this model (50).

Human evidence supporting epigenetic mechanisms of obesity

Certain genes may be particularly vulnerable to epigenetic changes. Metastable epialleles like the Avy allele in mice are subject to dramatic interindividual differences in methylation (37). Imprinted genes, for which the allele from 1 parent is normally silenced by methylation, are at risk of causing functional problems if the remaining copy is inappropriately silenced. Two rare human obesity syndromes, Prader-Willi and Beckwith-Wiedemann, can result from inappropriate methylation of imprinted genes (38). The search for metastable epialleles and imprinted genes affecting obesity in humans is of great interest.

Several studies have examined the effects of specific exposures on human fetuses or infants. A prospective study following 1,100 mother-child pairs from the prenatal period examined the effect of several perinatal exposures on obesity at age 3 years: prenatal smoking, excess weight gain during pregnancy, breast-feeding for fewer than 12 months, and poor infant sleep duration (51). This study found a progressive increase in risk of obesity with the addition of each risk factor (51). A meta-analysis of breast-feeding studies found a dose-response effect of breast-feeding duration on decreasing obesity prevalence. Each month of breast-feeding up to 9 months decreased the risk of obesity at 3 years by 4%, yielding an odds ratio of 0.68 for overweight in those breast-fed longer than 9 months compared with infants who were fed formula exclusively (52). These human studies did not include analysis of specific epigenetic markers for gene expression, but they suggest that early life exposures can have lasting effects on phenotype.

One study has demonstrated epigenetic changes in human growth-controlling genes associated with adverse perinatal events. The Dutch famine of 1944-1945 provides a unique opportunity to study humans exposed to well-defined undernutrition. Adults who were exposed to poor nutrition in utero had an increased prevalence of glucose intolerance, dyslipidemia, early coronary heart disease, and obesity (53,54). A recent study of adults who were exposed to the Dutch famine early in gestational development is reportedly the first to provide empiric support for the hypothesis that environmental exposures can cause epigenetic changes in humans. The insulin-like growth factor-2 (IGF-2) gene, which exerts control over fetal growth, is imprinted with the maternal allele normally silenced by methylation (55). Methylation of IGF-2 was decreased in adults who were exposed to famine early in utero compared with unaffected siblings and those exposed to famine only late in gestation. This was possibly an effect of decreased availability of methyl donors such as folate and the amino acid methionine during early development (43).

Implications of epigenetic mechanisms for obesity and public health

If epigenetic modifications that increase risk for obesity and chronic diseases occur widely in humans, the implications for public health interventions could be substantial. Epigenetic changes could underlie the increased risk of central obesity and cardiovascular disease among adults who experienced adverse in utero conditions, but at present direct evidence in humans is sparse. Epigenetic modifications have been proposed as contributors to variation in obesity across race, country, and immigrant status (35,56). Some prenatal and early life exposures such as smoking, inappropriate weight gain during pregnancy, gestational diabetes, and early feeding are known targets for intervention, but a modifiable biologic mechanism may provide an additional incentive to intervene early. Maternal stress or chemical exposures may be explored for epigenetic mechanisms affecting human disease. Research related to epigenetics draws on the expertise of multiple disciplines, including molecular biology/genetics, nutrition, environmental epidemiology, and life course epidemiology. An international scientific initiative, the Alliance for the Human Epigenome and Disease (AHEAD), has a goal to create a reference map of epigenetic modification sites (57). Prospective studies that collect biological samples early in life, such as the National Children's Study, may provide insight into epigenetic mechanisms of obesity and chronic disease (58).

Gut Microbiota

Differences in intestinal flora may explain some of the risk for obesity, yet like epigenetic mechanisms, evidence in humans is sparse. The gut microbiota consists of the microorganisms, predominantly bacteria, that inhabit the gastrointestinal tract. These microbes metabolize otherwise indigestible components of the diet, and the products of microbial metabolism affect the amount of energy absorbed (59). The combined gene pool of gut microorganisms enables absorption of simple sugars from complex polysaccharides and may influence fat storage by modifying lipoprotein lipase activity (60). RNA sequencing has been used to study bacterial species in stool samples (61). The species-level makeup of gut flora varies from person to person, but most analyses related to obesity have focused on how the relative proportion of 2 major bacterial divisions, Bacteroidetes and Firmicutes, differ between obese and lean people (62).

Animal studies of microbiota and obesity

The effect of microbiota on energy absorption and obesity has been demonstrated in mice. Germ-free mice have significantly less body fat than do mice with standard microbiota, and rapidly gain weight after inoculation (60). Obesity-prone mice, in contrast to their nonobese siblings, carry microbiota with an enhanced proportion of genes to break down polysaccharides, and their stools show increased fermentation products and decreased residual calories (59). When gut flora from obese or lean mice were transplanted to germ-free mice eating a high-fat, high-sugar diet, those receiving microbiota from obese mice gained significantly more fat than did those receiving lean mouse flora (59).

Human studies of microbiota and obesity

Thus far, human studies regarding the effect of intestinal flora have been observational and have had small sample sizes. A recent case-control study found gut flora in infancy predicted overweight later in childhood (63). Among children who were enrolled as newborns, 25 overweight 7-year-olds were matched with normal-weight controls for multiple factors including probiotics supplementation, antibiotic use, and breast-feeding duration (63). On average, overweight children had lower numbers of the genus Bifidobacterium spp. and higher numbers of Staphylococcus aureus in their stools during infancy (63). Bifidobacteria are the predominant flora of breast-fed infants and are hypothesized to affect weight gain through mucosal host-microbe crosstalk, immune regulation, and inflammation (64).

Other studies have examined the flora of obese adults. Studies of lean and obese twin pairs found a large variability among species of bacteria present, but a core group of functional genes was present across participants regardless of species type (65). In addition to this "core microbiome," the microbiomes of obese twins contained more genes involved in carbohydrate, lipid, and amino acid metabolism (65). A small study followed stool microbiota in 12 obese people randomized to low-fat or low-carbohydrate diets for a year. Obese participants had a smaller proportion of Bacteroidetes and a higher proportion of Firmicutes at baseline. During the study, the proportion of Bacteroidetes steadily increased as weight decreased on both diets, especially on the low-carbohydrate diet (66).

Potential implications of gut microbiota for obesity and public health

Other reviewers have suggested that from a public health perspective, it might be wise to avoid shifting too much focus from the known modifiable causes of obesity to gut flora (62). With that reasonable caveat, a small constant source of increased energy absorption could have a substantial effect on obesity in populations.

Throughout history, many cultures have used beneficial microorganisms to create fermented foods containing live microorganisms capable of modifying the makeup of intestinal flora, such as yogurt, curd, and kefir (62). Wide-ranging health claims have been made of commercially available probiotics (live nonpathogenic organisms) and prebiotics (fermentable substrates that enrich the gut for potentially beneficial organisms). Larger prospective studies are required to validate these health claims and to evaluate whether experimental changes to gut flora can affect obesity (67). The genetic sequencing techniques necessary to carry out this type of research are rapidly evolving (68). The National Institutes of Health has devoted substantial resources to the Human Microbiome Project to study the association of microbiota from the gut and other body sites with disease processes (69).

Conclusion

A systems approach to the obesity problem necessitates research that connects socioenvironmental factors with biological processes related to energy metabolism. By understanding how obesity results from the interaction of cellular factors with social factors, we can develop interventions that include molecular medicine and broad social policy. The potential biological drivers of obesity include evolutionarily conserved neurobiological mechanisms, epigenetic gene-environment interactions, and gut microbiota. These examples show the need for partnership among investigators across the spectrum of science. Research questions and hypotheses that are cross-disciplinary can aid development of interventions to prevent or control obesity at multiple levels.

HuangTTGlassTA30015200818111813Transforming research strategies for understanding and preventing obesityJAMA18854544LevinBE583Pt 22007425430Why some of us get fat and what we can do about itJ Physiol17584845LevinBE14Suppl 52006192S196SCentral regulation of energy homeostasis intelligent design: how to build the perfect survivorObesity (Silver Spring)17021365ZhengHBerthoudHR2008237583Neural systems controlling the drive to eat: mind versus metabolismPhysiology (Bethesda)18400690BerthoudHRMorrisonC2008595592The brain, appetite, and obesityAnnu Rev Psychol18154499LevinBEDunn-MeynellAA27812000R231R237Defense of body weight against chronic caloric restriction in obesity-prone and -resistant ratsAm J Physiol Regul Integr Comp Physiol10644644LevinBEKeeseyRE2742 Pt 21998R412R419Defense of differing body weight set points in diet-induced obese and resistant ratsAm J Physiol9486299AstrupAGøtzschePCvan de WerkenKRanneriesCToubroSRabenA696199911171122Meta-analysis of resting metabolic rate in formerly obese subjectsAm J Clin Nutr10357728WingRRHillJO200132334121Successful weight loss maintenanceAnnu Rev Nutr11375440LeibowitzSFHoebelBGBrayGABouchardC2007New York (NY)Informa Healthcare USABehavioral neuroscience and obesityHandbook of obesity: etiology and pathophysiology2nd edPliquettRUFührerDFalkSZyssetSvonCramonDYStumvollM3872006442446The effects of insulin on the central nervous system — focus on appetite regulationHorm Metab Res16933179KleinmanRE2008Elk Grove Village (IL)American Academy of PediatricsPediatric nutrition handbook6th edLevinBEDunn-MeynellAA28212002R46R54Defense of body weight depends on dietary composition and palatability in rats with diet-induced obesityAm J Physiol Regul Integr Comp Physiol11742822VerhagenJV5322007271286The neurocognitive bases of human multimodal food perception: consciousnessBrain Res Rev17027988AdamTCEpelES9142007449458Stress, eating and the reward systemPhysiol Behav17543357BerthoudHR2642002393428Multiple neural systems controlling food intake and body weightNeurosci Biobehav Rev12204189JamesWP26342008336352The epidemiology of obesity: the size of the problemJ Intern Med18312311TaheriS91112006881884The link between short sleep duration and obesity: we should recommend more sleep to prevent obesityArch Dis Child17056861CappuccioFPTaggartFMKandalaNBCurrieAPeileEStrangesS3152008619626Meta-analysis of short sleep duration and obesity in children and adultsSleep18517032TaverasEMRifas-ShimanSLOkenEGundersonEPGillmanMW16242008305311Short sleep duration in infancy and risk of childhood overweightArch Pediatr Adolesc Med18391138LandhuisCEPoultonRWelchDHancoxRJ12252008955960Childhood sleep time and long-term risk for obesity: a 32-year prospective birth cohort studyPediatrics18977973VanCauterEKnutsonKL159Suppl 12008S59S66Sleep and the epidemic of obesity in children and adultsEur J Endocrinol18719052MarshallNSGlozierNGrunsteinRR1242008289298Is sleep duration related to obesity? A critical review of the epidemiological evidenceSleep Med Rev18485764BlassEMAndersonDRKirkorianHLPempekTAPriceIKoleiniMF884-52006597604On the road to obesity: television viewing increases intake of high-density foodsPhysiol Behav16822530HalfordJCBoylandEJHughesGMStaceyLMcKeanSDoveyTM1192008897904Beyond-brand effect of television food advertisements on food choice in children: the effects of weight statusPublic Health Nutr18005487LewisMKHillAJ2231998206214Food advertising on British children's television: a content analysis and experimental study with nine-year oldsInt J Obes Relat Metab Disord9539187McClureSMLiJTomlinDCypertKSMontagueLMMontaguePR4422004379387Neural correlates of behavioral preference for culturally familiar drinksNeuron15473974ConnorSM1184200614781485Food-related advertising on preschool television: building brand recognition in young viewersPediatrics17015538ChouS-YRashadIGrossmanM5142008599618Fast-food restaurant advertising on television and its influence on childhood obesityJ Law EconHawkesC9711200719621973Regulating and litigating in the public interest: regulating food marketing to young people worldwide: trends and policy driversAm J Public Health17901436SingerJGMarketing ecosystems: framing brand management for business ecosystemsSan Diego (CA)CRM Systems IncAccessed January 15, 2009http://www.crm2day.com/library/50216.phpChesterJMontgomeryKInteractive food and beverage marketing: targeting children and youth in the digital ageAccessed January 15, 2009Berkeley (CA)Berkeley Media Studios Grouphttp://digitalads.org/documents/digiMarketingFull.pdfLoosRJLindgrenCMLiSWheelerEZhaoJHProkopenkoI4062008768775Common variants near MC4R are associated with fat mass, weight and risk of obesityNat Genet18454148FraylingTMTimpsonNJWeedonMNZegginiEFreathyRMLindgrenCM31658262007889894A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesityScience17434869KuzawaCWSweetE2112009215Epigenetics and the embodiment of race: developmental origins of US racial disparities in cardiovascular healthAm J Hum Biol18925573UbedaFWilkinsJF2008626101115Imprinted genes and human disease: an evolutionary perspectiveAdv Exp Med Biol18372794WaterlandRAMichelsKB200727363388Epigenetic epidemiology of the developmental origins hypothesisAnnu Rev Nutr17465856JirtleRLSkinnerMK842007253262Environmental epigenomics and disease susceptibilityNat Rev Genet17363974BarkerDJErikssonJGForsénTOsmondC316200212351239Fetal origins of adult disease: strength of effects and biological basisInt J Epidemiol12540728GluckmanPDHansonMACooperCThornburgKL359120086173Effect of in utero and early-life conditions on adult health and diseaseN Engl J Med18596274AllisCDJenuweinTReinbergD200723Cold Spring Harbor Laboratory PressCold Spring Harbor Laboratory PressEpigenetics17965594GodfreyKMLillycropKABurdgeGCGluckmanPDHansonMA615 Pt 220075R10REpigenetic mechanisms and the mismatch concept of the developmental origins of health and diseasePediatr ResHeijmansBTTobiEWSteinADPutterHBlauwGJSusserES1054420081704617049Persistent epigenetic differences associated with prenatal exposure to famine in humansProc Natl Acad Sci U S A18955703WaterlandRATravisanoMTahilianiKGRachedMTMirzaS329200813731379Methyl donor supplementation prevents transgenerational amplification of obesityInt J Obes (Lond)18626486DolinoyDCHuangDJirtleRL1043220071305613061Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early developmentProc Natl Acad Sci17670942LillycropKAPhillipsESJacksonAAHansonMABurdgeGC1356200513821386Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspringJ Nutr15930441RoemmichJNSmithJREpsteinLHLambiaseM159200723032310Stress reactivity and adiposity of youthObesity (Silver Spring)17890499MeaneyMJSzyfMSecklJR1372007269277Epigenetic mechanisms of perinatal programming of hypothalamic-pituitary-adrenal function and healthTrends Mol Med17544850ChampagneFA2932008386397Epigenetic mechanisms and the transgenerational effects of maternal careFront Neuroendocrinol 18462782OgrenMPLombrosoPJ4732008240244Epigenetics: behavioral influences on gene function, part I. Maternal behavior permanently affects adult behavior in offspringJ Am Acad Child Adolesc Psychiatry18512291GillmanMWRifas-ShimanSLKleinmanKOkenERich-EdwardsJWTaverasEM167200816511656Developmental origins of childhood overweight: potential public health impactObesity (Silver Spring)18451768HarderTBergmannRKallischniggGPlagemannA16252005397403Duration of breastfeeding and risk of overweight: a meta-analysisAm J Epidemiol16076830RoseboomTdeRooijSPainterR8282006485491The Dutch famine and its long-term consequences for adult healthEarly Hum Dev16876341RavelliGPSteinZASusserMW29571976349353Obesity in young men after famine exposure in utero and early infancyN Engl J Med934222HeudeBOngKKLubenRWarehamNJSandhuMS927200727342738Study of association between common variation in the insulin-like growth factor 2 gene and indices of obesity and body size in middle-aged men and womenJ Clin Endocrinol Metab17488802López-JaramilloPSilvaSYRodríguez-SalamancaNDurànAMosqueraWCastilloV1542008362372Are nutrition-induced epigenetic changes the link between socioeconomic pathology and cardiovascular diseases?Am J Ther18645341American Association for Cancer Research Human Epigenome Task ForceEuropean Union, Network of Excellence, Scientific Advisory Board45472052008711715Moving AHEAD with an international human epigenome projectNature18685699LandriganPJTrasandeLThorpeLEGwynnCLioyPJD'AltonME1185200621732186The National Children's Study: a 21-year prospective study of 100,000 American childrenPediatrics17079592TurnbaughPJLeyREMahowaldMAMagriniVMardisERGordonJI4447122200610271031An obesity-associated gut microbiome with increased capacity for energy harvestNature17183312BäckhedFDingHWangTHooperLVKohGYNagyA1014420041571815723The gut microbiota as an environmental factor that regulates fat storageProc Natl Acad Sci U S A15505215DiBaiseJKZhangHCrowellMDKrajmalnik-BrownRDeckerGARittmannBE8342008460469Gut microbiota and its possible relationship with obesityMayo Clin Proc18380992MaiVDraganovPV15120098185Recent advances and remaining gaps in our knowledge of associations between gut microbiota and human healthWorld J Gastroenterol19115471KalliomäkiMColladoMCSalminenSIsolauriE8732008534538Early differences in fecal microbiota composition in children may predict overweightAm J Clin Nutr18326589LeBouderERey-NoresJERabyACAffolterMVidalKThorntonCA1766200637423752Modulation of neonatal microbial recognition: TLR-mediated innate immune responses are specifically and differentially modulated by human milkJ Immunol16517743TurnbaughPJHamadyMYatsunenkoTCantarelBLDuncanALeyRE45772282009480484A core gut microbiome in obese and lean twinsNature19043404LeyRETurnbaughPJKleinSGordonJI4447122200610221023Microbial ecology: human gut microbes associated with obesityNature17183309TennysonCAFriedmanG1552008422427Microecology, obesity, and probioticsCurr Opin Endocrinol Diabetes Obes18769213FrankDNPaceNR2412008410Gastrointestinal microbiology enters the metagenomics eraCurr Opin Gastroenterol18043225TurnbaughPJLeyREHamadyMFraser-LiggettCMKnightRGordonJI44971642007804810The human microbiome projectNature17943116

The opinions expressed by authors contributing to this journal do not necessarily reflect the opinions of the US Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors’ affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above. URLs for nonfederal organizations are provided solely as a service to our users. URLs do not constitute an endorsement of any organization by CDC or the federal government, and none should be inferred. CDC is not responsible for the content of Web pages found at these URLs.

Suggested citation for this article: Haemer MA, Huang TT, Daniels SR. The effect of neurohormonal factors, epigenetic factors, and gut microbiota on risk of obesity. Prev Chronic Dis 2009;6(3). http://www.cdc.gov/pcd/issues/2009/jul/09_0011.htm. Accessed [date].