Skip to main content
Endocrine Reviews logoLink to Endocrine Reviews
. 2023 Nov 24;45(2):253–280. doi: 10.1210/endrev/bnad031

Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk

Alicja A Skowronski 1,2, Rudolph L Leibel 3,4,, Charles A LeDuc 5,6
PMCID: PMC10911958  PMID: 37971140

Abstract

This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS—including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.

Keywords: obesity, adiposity, developmental programming, hypothalamus, epigenetics

Graphical Abstract

Graphical Abstract.

Graphical Abstract


Essential Points.

  • Maternal obesity has become more prevalent in recent decades and can alter lifetime obesity risk in successive generations of progeny

  • Mouse studies indicate that overnutrition of pups can influence the development of hypothalamic, brain stem, and hedonic circuits critical in energy homeostasis

  • Various hormones—including leptin, insulin, ghrelin, amylin, and glucagon-like peptide-1—may have direct effects on adult adiposity via the effects on developing feeding circuits

  • The nutritional environment during development may be conveyed to progeny through epigenetic marks which can be vertically transmitted to subsequent generations

  • Experiments investigating the impact of nutritional environment during early postnatal brain development in mice are likely relevant to late gestation in humans

The prevalence of childhood obesity has increased dramatically over the past 4 decades; its prevalence is currently 18.5% for children between 2 and 19 years of age (1). The long-term health consequences of obesity—increased risk of developing cardiovascular disease, type 2 diabetes (T2D), stroke, and some cancers—represent staggering current and projected burdens on the economy and society (2-4). A newly revealed complication of obesity and its negative effects on the global health care system is that obesity is a significant risk factor for severe disease and complications resulting from SARS-CoV-2 infection (5). In addition, the implementation of lockdown measures during the global COVID-19 pandemic (ie, school and businesses closures) aimed at halting the spread of SARS-CoV-2 has led to increased weight gain in adults (6-8) and children (9, 10) by increasing sedentary behaviors and changing eating habits (11, 12). Increased adiposity in children puts them at higher risk of obesity in adulthood. Since maternal obesity per se (as distinct from shared genotype) increases the risk of obesity in offspring, the Covid-19 pandemic will likely affect obesity prevalence in progeny of these gestations, and possibly in subsequent generations. In addition to imposed economic burdens, obesity has profound effects on quality of life as well as life expectancy (13), especially in younger adults (14, 15). Individuals with severe obesity have an estimated 5- to 20-year reduction in life expectancy (14). Obese adolescents are 22-fold more likely to be obese as adults and efficacious obesity treatments are limited; therefore, prevention of early obesity is critical to reduce the disease burden of this epidemic (16, 17).

Perinatal Programming of Body Weight: Evidence From Humans

Concomitant with large increases in obesity prevalence generally, maternal obesity has become common and could have long-lasting impacts on future generations. Genetic predisposition is an important contributor to obesity risk. Heritability studies suggest that ∼40% to 70% of the variation in adiposity among individuals within a shared environment reflects genetic factors (18-20). Body mass index (BMI)–associated genetic signals are highly enriched for variants that are in or near genes expressed in the central nervous system (CNS) (21, 22). With the advent of ’ ’omics level genetic susceptibility scoring, relative obesity propensity can be accurately scored using 2.1 million single nucleotide polymorphisms (23). These polygenic risk scores have been used to stratify populations to isolate the effects of single gene mutations on body weight (24), coronary heart disease, and breast cancer (25), as well as to identify genetic indicators of response to pharmaceutical interventions (26).

The rapid increase in obesity during the past 50 years cannot be explained by gene sequence changes. Transmission of altered methylation/acetylation gene signatures—in response to environmental influences—is a possibility discussed below. In many countries, the current environment is highly obesogenic by virtue of a combination of readily available highly palatable and energy-dense foods and sedentary lifestyles. While there is a lack of support for significant declines in physical activity in the last 4 decades (27) and it is difficult to accurately measure energy intake, 1 or both of these factors must be driving secular increases in obesity. Epidemiological studies as well as mouse models suggest that obesity and metabolic disease originate not only from genetic and environmental factors, but also from their interplay with the developmental milieu. Identifying mechanisms possibly linking the perinatal environment to future health outcomes is important to the prevention of obesity and obesity-related diseases.

The Dutch Hunger Winter of 1944-45 provided a tragic opportunity to investigate the impact in adulthood of gestational undernutrition on disease risk. Fetuses exposed to famine during the first 2 trimesters of gestation had a higher incidence of obesity and cardiovascular disease in adulthood (28). Undernutrition during late gestation resulted in lower birth weight and those individuals continued to have lower rates of obesity (28, 29). Subsequently, David Barker proposed the “Fetal Origins of Adult Disease” hypothesis in which he postulated that adverse or suboptimal perinatal developmental environment leads to increased risk of disease in adult life. Barker emphasized a relationship between low birth weight and hypertension and coronary heart disease risk in adulthood (30); subsequently, numerous epidemiological studies have demonstrated that low birth weight is associated with increased risk of metabolic disease (reviewed in (31)). As a mechanism, Barker's “thrifty phenotype” hypothesis proposed that developmental programing by undernutrition primes the organism for low energy intake through metabolic adaptations that maximize survival in food-scarce environments (32). However, in an environment in which access to high-energy food is plentiful and easily accessible, the anticipatory adaptations can become detrimental to the individual's health.

At the time of Barker's observations, investigators were focused on the influence of poor nutritional state, stress, or disease of the mother on the outcomes in progeny as adults. However, over the intervening 3 decades, as populations have become more obese, focus has shifted towards developmental programming resulting from maternal overnutrition (33-35). There is currently more interest in the effects of excess materno-fetal nutrients on the long-term consequences in the offspring. Such developmental programming is the focus of “Developmental origins of health and disease” (36), predicated on the idea that environmental influences during critical periods of development and growth (which includes the gestational period through early postnatal development) may have adverse consequences for an individual's future health. These consequences presumably reflect both static structural changes in the brain, islets, adipose tissue and other organs as well as “imprinting” of molecular/biochemical pathways mediating a range of physiological processes such as blood pressure and lipid homeostasis. But the precise nature and mechanism of these putative effects remain poorly understood.

Prepregnancy obesity in US women increased from 26.1% to 29% between 2016 and 2019 (37). Excess adiposity leads not only to adverse pregnancy outcomes (38) (eg, increases in maternal mortality (39), increased risk of miscarriage (40), gestational hypertension (41), preeclampsia (41-43), gestational diabetes (GDM) (41, 44), stillbirth (43), and cholecystitis (45)), but also imperils the developing child (including large for gestational age [LGA] babies, fetal defects, congenital anomalies and perinatal death) (46). GDM, hypertension, and preeclampsia contribute to a suboptimal intrauterine environment. Maternal obesity (“controlling” for shared genotype) predisposes offspring to obesity, diabetes, hypertension, and cardiovascular disease (47-49). Infants born to obese mothers are more likely to be LGA (birth weight >90th percentile for gestational age and sex) or macrosomic (birth weight >4000 g) (43, 50, 51), which puts them at a higher risk of obesity later in life (52, 53). Obesity of both parents increases the risk of obesity in the offspring (54), but maternal pregravid BMI is the strongest predictor of childhood obesity (55); maternal obesity is a stronger determinant than paternal obesity of progeny neonatal BMI (56), as well as offspring BMI at 2-3 (56), 5-7 (57), and 14 (33) years of age. Excessive weight gain (based on Institute of Medicine gestational weight gain recommendations) during pregnancy is associated with higher infant adiposity (58, 59). Infants of prepregnancy obese women have the highest fat mass regardless of degree of maternal weight gain during gestation (59).

LGA babies have increased risk of obesity in adulthood (60, 61), and rapid weight gain in infancy is associated with later risks of obesity and metabolic disease, particularly insulin resistance (IR) (62, 63). A meta-analysis of 66 studies including 643 902 individuals aged between 1 and 75 years found that infants born at high birth weight (>4000 g) are at higher risk of overweight in adulthood (odds ratio [OR] 1.66). High rates of postnatal weight gain, termed “catch-up” growth, often occur in small for gestational age (SGA) infants. A systemic review of data reported from 1927 to 1994 found that birth size (infants at the highest end of the distribution for weight or BMI) and growth rate during infancy (babies who grow rapidly) are related to risk of obesity in children and adults (64). Rapid infant growth correlated with higher rates of obesity in childhood, adolescence, and early adulthood (64). A subsequent Finnish study that correlated postnatal weight and height velocity from birth to 2 years of age with metabolic outcomes at 31 years of age found that higher peak weight velocity was associated with increased incidence of adult obesity (by BMI and weight circumference) and hypertension, while peak height velocity correlated with blood pressure and weight circumference (65). A large-scale population-based study in Germany found that the most rapid BMI acceleration in obese adolescents occurred between 2 and 6 years of age and that 90% of obese 3 year olds remained overweight or obese in adolescence (66). These longitudinal studies clearly point to obesity in early childhood being an important predictor of obesity in adolescence or adulthood (67). A systematic review of over 200 000 children followed to adulthood showed that obese children and adolescents were around 5 times more likely to be obese in adulthood than those who were not obese (68). A Mendelian randomization study found that genetic predisposition to higher childhood BMI (study used 15 single nucleotide polymorphisms (associated specifically with childhood rather than adult obesity to test effects that are specific to early-life BMI) was associated with increased adult BMI (the effect size: 1 SD increase in childhood BMI associated with a 0.587 SD increase in adulthood BMI) (69). Importantly, Mendelian randomization studies demonstrate that childhood obesity (controlled to some extent for genetic risk) has a causal association with adult T2D (OR up to 1.83) and adult coronary heart disease (OR 1.38) (69, 70), suggesting that adult disease is driven by childhood obesity independently of genetic obesity risk.

Diabetes (type 1 diabetes [T1D], T2D, and gestational [GDM] diabetes) and impaired glucose tolerance during pregnancy have been linked to increased infant birth weight and more specifically, infant adiposity (71-74). Infants of diabetic mothers are born LGA and show a higher BMI throughout childhood (75) and adulthood (76). Intense treatment of GDM reduces the likelihood of LGA infants (77, 78) and tight glycemic control in gravidas with T1D was shown to reduce the incidence of LGA in some studies, but others indicate that even with well controlled maternal glucose, infants of mothers with T1D are at increased risk of macrosomia (79, 80). The precise glycemic targets required to mitigate LGA are debated, and may differ depending on the type of diabetes (81). In women with GDM, pregravid obese or overweight BMI increased risk of macrosomic baby even when hyperglycemia was well controlled with diet (82). In overweight or obese women with well-controlled GDM treated with insulin, the macrosomia risk was comparable to normal weight gravida (82). It is unclear by what mechanisms maternal insulin treatment may decrease neonatal macrosomia rates other than its effects on glycemia. In Pima Indian women who gave birth to children before and after a T2DM diagnosis the sibling who was exposed to a diabetic intrauterine environment had a greater risk of obesity and diabetes than offspring born before the mother was diagnosed with diabetes (83). While this study found no increase in diabetes propensity associated with paternal diabetes status (83) there have been other studies reporting that paternal obesity and diabetes status can influence susceptibility to future metabolic disorders in offspring (84). These data indicate that in addition to the genetic predisposition, intrauterine exposure to hyperglycemia (and attendant metabolic changes) per se contributes to the risk of obesity in the offspring.

GDM is often associated with maternal obesity and characterized by hyperglycemia and hyperinsulinemia. While insulin does not cross the placenta (85, 86), women with diabetes have increased circulating glucose concentrations leading to higher transplacental glucose transfer. Increased fetal glucose concentration stimulates fetal insulin production leading to hyperinsulinemia and fetal overgrowth (87). The Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study of 23 316 mother–newborn pairs found a linear relationship between increased maternal blood glucose measured at 24 to 32 weeks of gestation and cord blood C-peptide and an association of each of these parameters with birth weight (88).

Progeny of women with prepregnancy diabetes are at an increased risk for congenital malformations (organ teratogenesis) with some studies reporting up to 10-fold greater risk (89-91). The highest occurrence of birth defects associated with prepregnancy diabetes is congenital heart disease but other, noncardiac malformations include nervous system (ie, neural tube defects), respiratory, skeletal tissue, and urinary tract abnormalities (91, 92). Compared with pre-existing diabetes, GDM (usually diagnosed in the third trimester) is associated with fewer birth defects with weaker associations since most birth defects develop in the first trimester. Freinkel hypothesized that abnormal fuel levels (not limited to glucose) during gestation can not only induce congenital malformations but also have far-reaching implications on offspring physical and behavioral development (93). He coined the term “fuel-mediated teratogenesis,” proposing that the maternal fuels (in diabetes: elevated glucose, amino acids, ketones, and lipids) influence the development of metabolic tissues including pancreatic beta-cells, adipose tissue, and CNS cells undergoing differentiation, proliferation, and maturation in utero. It is possible that structural changes in the CNS resulting from gestational exposure to these abnormal fuel levels cause increased body weight throughout life, while higher fat mass at birth does not mediate adult obesity but could serve as an early marker of future obesity.

Direct administration of insulin via minipumps to fetal rhesus monkeys for 3 weeks prior to birth increased birth weight (by 23-27%), and increased weights of the placenta, heart, liver, and spleen, while not affecting the mass of other organs including brain and skeletal muscle (94). Similarly, a single insulin injection into a fetal rat at embryonic day 18.5 was sufficient to increase in body weight by 7% to 12% (95, 96); this increase persisted for the first 2 weeks of life (95). However, supplementing insulin to the pregnant rat dam (also through subcutaneous minipump from day 14 of gestation until parturition), resulted in intrauterine growth retardation (IUGR) of the fetuses, presumably due to decreased availability of metabolic substrates for growth (dams were hypoglycemic due to hyperinsulinemia with no difference in weight gain compared to control dams from baseline). The pups were hypoglycemic and hypoinsulinemic during gestation and after birth (97). These data suggest that fetal hyperinsulinemia is the primary driver of fetal overgrowth. Consistently, infants of diabetic or hyperglycemic mothers, display elevated cord blood insulin (88), and this fetal hyperinsulinemia is strongly associated with macrosomia at birth (88, 98, 99). A heterozygous loss of function mutation in glucokinase (GCK)—the most common cause of maturity-onset diabetes of the young (MODY)—is characterized by mild hyperglycemia resulting from a defect in glucose sensing by beta cells. Individuals with GCK-MODY have reduced concentration of insulin at any given glucose concentration (100). Gravidas with GCK-MODY have mildly elevated glucose but the body weight outcome in the offspring is dependent on the offspring genotype: the sibling who does not inherit the GCK mutation has a higher birthweight than a sibling with the mutation presumably due to decreased fetal insulin secretion in utero in the sibling with the GCK mutation. Offspring with paternally transmitted GCK mutations born to an unaffected mother have reduced birthweight due to intrauterine hypoinsulinemia (101).

Hyperinsulinemia and IR in gravidas with GDM has been linked to increased placental growth during early pregnancy (102, 103), while fetal hyperinsulinemia is thought to drive the increased placental growth (104) in later pregnancy which is consistent with higher expression of insulin receptors (InsRs) on the maternal side of the placenta early on and a shift of InsR expression to fetal side in late gestation (105). In offspring born to mothers with GCK-MODY, those with GCK mutation had reduced placental weight compared with those offspring without the mutation. These results implicate a role for insulin in placental growth. Fetal hyperinsulinemia increases placental vascularization (106), which is a mechanism of placental adaptation to metabolic stress.

Animal studies of in utero insulin administration indicate insulin's direct role in fetal growth. In humans, since obesity in gravidas is associated with LGA infants, and maternal obesity is associated with T2D and GDM in the gravida, it is challenging to determine the degree to which the intrauterine hyperglycemia per se drives increased fetal growth independently of other aspects associated with the maternal obese state. Studies controlling for maternal prepregnancy BMI found that the offspring of mothers with GDM had an increased incidence of overweight throughout childhood (2, 8, and 11 years of age (107)) compared with offspring of mothers with T1D or controls without diabetes; maternal obesity was a strong predictor of overweight in children (107). These studies highlight the importance of maternal and fetal hyperinsulinemia in predisposing to later obesity in progeny.

While disentangling genetic from development–environmental contributions to obesity outcomes is challenging—particularly in humans—weight gain between successive pregnancies is associated with an increased risk of LGA neonates, while weight loss after an LGA birth is correlated with a lower risk LGA in the next pregnancy (108-111). Additional evidence implicating developmental environment rather than shared genetic factors in programming of obesity comes from women who had children both before and after bariatric surgery. Obesity is reduced in offspring (aged 2-26 years) born after maternal bariatric surgery relative to siblings born prior to surgery (112-114), and the latter progeny show improved cardiometabolic markers into adolescence (113). A retrospective, Australian population-based study compared birthweight outcomes in women who had bariatric surgery between first and second pregnancy and showed reduced rates of LGA infants (OR 0.63) in the second pregnancy (115). These data suggest that the developmental environment contributes directly to increased offspring obesity. Differential gene methylation is a potential mechanism mediating these obesity differences (116).

The underlying cellular and molecular mechanism(s) for effects of maternal “programming” are unclear. Because human data cannot directly address these questions—all such influences are implied by epidemiological evidence and this evidence is confounded by heterogeneous genetics, diet, and lifestyle—animal models of identical genetic background and environmental exposure are relied on. Genetically modified animals have been indispensable in broadening our understanding of molecular mechanisms underpinning the developmental programming of body weight.

Rodent Models of Developmental Programming

Numerous investigations of the effects of nutritional status during early development on energy homeostasis in rodents have confirmed that gross physiological phenotypes are generally consistent—allowing for differences in the timing of neurodevelopment—between humans and rodent models. This correspondence is, of course, critical to the utility of rodent models use in the investigation of the molecular mechanisms by which early development programs adult body weight.

Perinatal Underfeeding

Rodent models of perinatal undernutrition primarily include maternal total calorie restriction (caloric intake usually decreased by 30-70%) or protein restriction (isocaloric diet containing 8-10% kcals from protein vs 20% in normal chow) during gestation and/or lactation (117-122), and increasing litter size to reduce breast milk availability per offspring (123, 124) (Fig. 1). Maternal calorie or protein restriction during gestation (durations differ from 1 to 2 weeks to the entire 21-day pregnancy) leads to significantly lower body weight of fetuses at birth (IUGR) than rat dams fed ad libitum (125). IUGR is associated with metabolic phenotypes related to glucose-insulin homeostasis including beta cell dysfunction, IR, and glucose intolerance in adult rodent offspring (126-130) (Fig. 1).

Figure 1.

Figure 1.

Effects of diet composition and caloric underfeeding and overfeeding during gestation and/or lactation on the programming of body weight in the offspring. Note that different conditions may lead to similar outcomes as indicated by the arrows pointing to the effect boxes. Abbreviations: IUGR, in utero growth restriction; BW, body weight; DIO, diet-induced obesity; HFD, high-fat diet. Figure created with BioRender.com.

The timing of maternal calorie restriction is critical, as it exerts differential effects on the offspring, and in some models the effects are sexually dimorphic in rats (117, 131) and mice (132). Both male and female rat offspring born to dams undernourished only during pregnancy are smaller at birth than controls. When these rat pups are cross-fostered to dams fed ad libitum they gain weight rapidly (“catch-up” growth) and most studies report increased body weight and fat percentage vs control offspring in adulthood when maintained on a regular chow (117, 118). This catch-up growth is associated with increased susceptibility to diet-induced obesity reported in both mice (122) and rats (133). When maternal undernutrition spans from gestation through the lactation period, the accelerated postnatal growth is prevented resulting in ad libitum-fed offspring that have persistently lower body weight than control animals found in both mice (132) and rats (117, 118, 134). Howie et al reported that under these circumstances female, but not male, rat offspring have a reduction in percentage of body fat at 5 months of age (117). When maternal undernutrition is restricted to the lactation period only, mouse (132) and rat (134) offspring have persistently reduced body weight and fat when subsequently fed regular chow ad libitum. Howie et al found such differences only in female rat offspring (117). Desai et al exposed the progeny nursed by undernourished rat dams to a Western-style diet in adulthood. There was no difference in body weight of the rats compared with controls but males showed a small increase in body fat percent (difference not seen in females) (135).

Similar to maternal calorie restriction, restricting the amount of protein in maternal diet in rats during gestation is sufficient to induce fetal growth retardation (120). Providing these pups with proper postnatal nutrition, either by cross-fostering them with dams fed control diet and/or placing them in smaller litters (for increased milk availability), results in “catch-up” growth, but the resulting pups are more susceptible to diet-induced obesity (reports of consistent effects in both rats and mice) (133, 136-138). When a low-protein diet is enforced throughout rat dam pregnancy and lactation, the offspring remain significantly lower in weight throughout life compared to controls (133, 139, 140). In fact, nursing by dams fed a reduced protein diet only in postnatal life results in reduced preweaning body weight that persists into adulthood found in mice (136) and rats (120, 140, 141).

While there are some inconsistencies among studies assessing the effects of gestational and/or postnatal calorie and protein restriction on body weight in the adult offspring, this is not unexpected given variations in the experimental conditions (including the exact timing) and differences in rodent strains and whether mice or rats were studied. The most consistent phenotype is evident in the offspring that experience intrauterine growth restriction (via caloric or protein restriction of the dam) followed by “catch up” growth during lactation (via cross-fostering to ad libitum-fed dams) constituting a mismatch between the prenatal and postnatal environments. These rats are more prone to weight gain when exposed to hypercaloric (32% fat and 10% sucrose liquid) diet as adults (133). Some reported consequences of prenatal undernutrition that may contribute to subsequent somatic phenotypes include alterations in hypothalamic expression of energy homeostasis genes (142), persistent changes in the plasma metabolome (143), decreased sensitivity to leptin (144, 145), and disturbances in the hypothalamic “wiring” of the proopiomelanocortin (POMC) neurons (121), all reported in rat offspring. Interestingly, the offspring of rat dams that were calorie restricted during gestation and lactation experienced a delay in postnatal leptin surge (more details on the surge in the following sections) (121, 146), whereas those that were born to gestationally calorie-restricted mouse dams showed an augmented and earlier leptin surge, likely a result of accelerated postnatal growth of fat mass (122).

To define the effects of undernutrition on developmental programming of body weight specifically during the lactation period, the amount of milk available per pup is experimentally reduced by expansion of litter size. Offspring raised in large litters experience slower growth and are significantly lower in body weight (up to 35% by weaning) during nursing period compared to offspring reared in normal size litter, a consistent phenotype reported in both mice and rats (124, 147-150). Most studies report that the mouse (123, 148) and rat (147, 151) offspring remain smaller in adulthood, while some find that the offspring display catch-up growth after weaning and reach higher adiposity than offspring reared in normal size litters. Undernutrition (by increasing pups in a litter) results in a delayed postnatal leptin surge of decreased magnitude reported in mice and rats (121, 124, 149) and altered expression patterns of hypothalamic genes involved in the melanocortin axis found in mice (147).

Perinatal Overfeeding

Rodent models of perinatal overfeeding include maternal obesity induced by variations of high-fat diet (HFD) (some of which include sugar) during gestation and/or lactation as well as overnutrition during nursing by litter size reduction. These studies generally suggest that perinatal overnutrition results in obesity and metabolic abnormalities (such as IR or hepatic steatosis) in offspring as adults (Fig. 1).

Increased body weight/adiposity in rodent adult offspring is one of the most consistently associated phenotypes in experiments of imposed maternal obesity during perinatal period. Not surprisingly, characteristic metabolic phenotypes are often associated with obesity in these models, including disturbances in glucose tolerance (in mice and rats) (152, 153), IR (in rats) (153), pancreatic b-cell dysfunction, fatty liver (in mice) (154, 155), increased in systolic (in mice) (155, 156) and diastolic blood pressure (in mice) (152), and decreased hypothalamic leptin and insulin signaling (in rats) (153, 157). Rodent studies suggest that maternal obesity/overnutrition during both prenatal and postnatal (lactation) periods are important in programming body weight in the offspring. Some mouse studies report that maternal HFD-feeding during gestation or lactation each contribute independently to increased adiposity of the offspring, and that exposure during both periods produces even greater increase in adiposity in the adult offspring (152, 156). Others report that high-energy density diets (this study used a “junk” also known as “cafeteria” diets biscuits, marshmallows, cheese, jam doughnuts, chocolate chip muffins, butter flapjacks, potato crisps, and caramel/chocolate bars) during both gestation and lactation periods are necessary to program increased weight and propensity for high-density foods in the adult rat offspring (158). However, limiting maternal HFD exposure to lactation alone may be sufficient and has a greater influence on weight of the adult offspring than gestational maternal HFD found in rats and mice (159, 160). A recent study investigating the impact of increasing levels of fat content in maternal diet (during lactation only) on mouse offspring adiposity and found that ≥41.7% dietary fat compared with 8.3% or 25% resulted in persistent increases in offspring body weight at weaning and at 14 weeks of age when maintained on regular chow (161, 162). HFD (41.7% fat) exposure of 14-week-old mouse offspring nursed by dams fed HFD with fat content ≥41.7% resulted in an increased susceptibility to weight gain which was more pronounced in males than females; interestingly, increasing fat in the maternal diet to 58% or 66.1% had little or no additional impact (161, 162), suggesting that 41.7% of fat in the maternal diet is sufficient to induce the body weight changes in the offspring (Fig. 1).

In the rodent model of postnatal overfeeding via litter size reduction, the remaining pups (eg, n = 3) have greater caloric availability during the suckling period causing an increased growth rate compared with pups reared in normal size (eg, n = 8) litter. The inverse relationship between litter size and pup body weight throughout suckling suggests that in the early postnatal period, immature pups lack satiety signals and do not regulate food intake but rather the amount of ingested milk is solely dependent on availability (163). In fact, a major regulator of energy homeostasis, leptin, does not suppress food intake or body weight in neonatal mice and rats (164-166). In addition to increased weight gain during the lactation period, mouse pups raised in small litters have persistently higher body weights throughout life (up to 23 weeks of age on chow diet) compared to controls and gain more weight when exposed to HFD at 6 weeks of age (167, 168).

While the models of reduced litter size and maternal obesity/HFD feeding differ, primarily in metabolic status of the nursing dams, both of them result in chemical compositional changes to the breastmilk. Rat dams that are fed a high-energy density diet show an increase in caloric density of the breastmilk (∼20% increase on P21 based on calculation from proportions of macronutrients (169)) and elevation of macronutrient concentrations including sugar, protein, triglycerides and cholesterol (153, 157, 169, 170). The data on effects of maternal HFD feeding on milk production are conflicting. Del Prado et al found the volume of milk production to be higher (171) while Bautista et al reported less milk volume per day (170) in HFD-fed compared to control rat dams. Maternal cafeteria feeding (consisting of salami, cheese crackers, and chocolate chip cookies) during different time combinations, including before conception, and during gestation and lactation, alters the composition of milk produced. The largest effect of maternal diet on milk composition in rats was during lactation (172). In cafeteria-fed rat dams, the milk fat content was increased and consequently the caloric density calculated from macronutrient composition was higher (172). Interestingly, even when the diet of dams nursing normal or reduced number of rat pups is the same, in dams with reduced size litter the fat content of milk is elevated while protein and sugar concentrations are largely unchanged (173-175). Caloric density of milk from rat dams nursing small litters was higher by up to 30% (174, 176), indicating that in addition to increased milk volume consumption the increased weight/fat gain of the rat pups reared in reduced-size litters is a result of the increased fat content and caloric density of the breastmilk (174).

Rat and mouse pups experience a leptin surge during which concentrations of circulating leptin are elevated 5- to 10-fold with peak concentrations occurring between postnatal day 7 and 10; these concentrations are independent of fat mass (177). We have since shown that the leptin concentrations in neonatal mice are higher than what would be predicted by their fat mass up to postnatal day 27 (149). While it is unclear by what mechanism this postnatal leptin surge is induced, the nutritional status of the pups during this period affects the timing and magnitude of the leptin surge. In both models of overnutrition described above—either the reduced litter size or rearing by dams fed HFD during gestation and/or lactation in both mice and rats—the leptin surge in the pups starts earlier with an increased magnitude (124, 149, 156, 178) even when plasma leptin concentrations are adjusted for fat mass (149).

The reported effect sizes on body weight in adult offspring of obese dams and small litter size overfeeding are variable. The phenotypic inconsistencies likely result from different experimental design including the timing of HFD feeding or overnutrition. Similar to perinatal undernutrition, the design of overnutrition experiments varies widely across reported studies. These variables include the timing of hypercaloric diet exposure (pregestation, gestation, lactation, or all 3), the diet used (different % of calories coming from fat in HFDs, and different proportions of sucrose), sex of studied offspring, the strain of mice or rats used, and whether the offspring were cross-fostered (this is especially important when the dam was switched from HFD during pregnancy to chow at parturition). Cross-fostering is critical to avoid the effects of postnatal undernutrition which would occur as a result of food intake decrease following a HFD to chow diet switch [179).

Mechanisms Mediating the Developmental Programming of Adiposity

Hypothalamic Development

The arcuate nucleus of the hypothalamus (ARH) is adjacent to the third ventricle and median eminence, a circumventricular organ, all located within the mediobasal hypothalamus with a “permissive” blood–brain barrier access. The 2 best characterized populations of leptin-responsive neurons—POMC and neuropeptide Y (NPY)/agouti-related protein (AgRP)—reside in the ARH and are therefore well positioned to relay circulating peripheral signals of energy homeostasis to other parts of the hypothalamus and CNS. Hypothalamic cells and circuits are sensitive to the impact of obesity and HFD-feeding during development. In rodent models, these effects are conveyed in part by circulating molecules such as leptin, insulin, ghrelin, and amylin. Perturbations in the concentrations of these molecules affect neurogenesis of feeding-critical neurons generated during gestation, axonal outgrowth of neurons participating in feeding circuits during early postnatal period, and/or epigenetic modifications to genes involved in energy homeostasis (180, 181). The pathways predicted to be primarily affected by such perinatal programming are those critical in energy homeostasis, such as those involving leptin- and insulin-responsive POMC and AgRP neurons (182, 183).

With the important caveat that mice are born earlier in neural development than humans, hypothalamic development in rodents begins during gestation and extends well into the postnatal period (Fig. 2). Rodent timing of neurogenesis of hypothalamic neurons, using 3H thymidine labeling, suggests that the majority of neurons are created between E11 and E14 with some differences among nuclei (187, 188). In a more recent study, Ishii and Bouret used bromodeoxyuridine to systematically identify neurogenesis in the hypothalamus, specifically those neurons that are a part of leptin-sensitive circuits. They found that there is a sharp peak of neurogenesis for leptin-responsive hypothalamic neurons that occurs at E12 and that the majority of these neurons are generated between E12 and E16 (189). Expression of Lepr (leptin receptor) is detected in the developing rodent brain at E10.5 (measured in whole brain with reverse transcription polymerase chain reaction); using in situ hybridization, Lepr mRNA expression is observed in the premamillary hypothalamic nucleus from E16.5 and in the ARH and ventromedial hypothalamic nucleus at E18.5 (190). Maternal HFD feeding during pregnancy increases expression of POMC and NPY neuropeptides in E18 fetal whole brains (191). Leptin influences neurogenesis during gestation (192) and lactation (193) in mice. Congenitally leptin-deficient, Lepob/ob, mice have smaller brains (194) with fewer neurons at E16 and E18, and fewer recently created neuroepithelial cells at E14 and E16 (192). The number of these neuroepithelial cells in E16 Lepob/ob embryos is normalized by intracerebroventricular leptin injection at E14 (192). Leptin induces neuronal proliferation and differentiation in ex vivo hypothalamic neurospheres (derived from the embryonic E20 rats) in a dose-dependent response (195). Neurogenesis of the feeding circuit neurons in mice is thought to be confined primarily to E12-E16. However, in Lepob/ob mice, systemic leptin injections for 2 weeks starting at week 4 of age (just after weaning) increases brain mass and total brain DNA content at 6 weeks of age. In 9 weeks old Lepob/ob mice, such injections do not affect brain mass or DNA content at 11 weeks of age (193). Clearly, there are important developmental windows/critical periods for effects of “environmental” influences on virtually all aspects of brain development.

Figure 2.

Figure 2.

Comparison of brain development in humans (top) and rodents (bottom) from gestation to maturity. Humans and rodents experience similarly sequenced brain maturation but the timing is shifted to the right in rodents. At birth, humans have more mature brains than mice (by timing of neural events such as neurogenesis, synaptogenesis, and myelination (184), morphological observations (185), and transcriptional analysis (186)). In humans the peak brain growth rate occurs in the last 4 weeks of gestation but in mice this peak occurs in the second postnatal week. Figure created with BioRender.com.

In mice in early development, between E11.5 and 13.5, Pomc is expressed broadly across the hypothalamus but, by adulthood, only 50% of these neurons remain Pomc positive while the remainder switch fates, including differentiating into NPY/AgRP neurons (196). Offspring of mice fed HFD, in addition to maternal IR (IR dams were heterozygous for a null allele of Insr and displayed hyperinsulinemia with normal glucose concentrations), showed a transient increase in body weight between 4 and 8 weeks of age when fed HFD after weaning. The offspring of HFD-fed IR dams had increased numbers of POMC-expressing cells in the ARH at P9 indicating that ambient insulin concentration in the pup gestational environment can alter the switch from POMC to NPY/AgRP neurons (197). These data suggest that midgestation is an important developmental period during which the hypothalamus may be particularly sensitive to aberrant intrauterine environmental factors which could affect neurogenesis.

In rodents, hypothalamic neurons created in midgestation do not complete neuronal circuits or send axonal projections to their terminal target sites until the postnatal period (198, 199). Immaturity of feeding circuits during the first 1-2 weeks of postnatal life accounts for the absence of a difference in adiposity of leptin deficient or leptin overexpressing mice prior to P10. At birth, neural projections originating from the ARH are immature and are established primarily within the first 2 weeks of postnatal life (199), processes that are concurrent with the naturally occurring leptin surge (149, 177). In rodents, plasma leptin concentrations increase 5- to 10-fold in the second week of life, and peak at P10; this surge persists for the first 4 postnatal weeks (149, 177). During this period, leptin acts as a neurotrophic factor to stimulate growth of axonal projections of the AgRP/NPY neurons (198-200). Axonal innervation patterns are exquisitely timed in the mouse. Mature innervation from the ARH to the dorsomedial hypothalamic nucleus (DMH) is achieved by P6, then to the periventricular hypothalamic nucleus (PVH) between P8-10, followed by projections from ARH to lateral hypohtalamic area (LHA) by P12 (199). Congenitally leptin-deficient, Lepob/ob, mice display severe reduction in axonal projection density from AgRP neurons in the ARH to the PVH, DMH, and LHA, which persists through adulthood (198). These diminished ARH projections in Lepob/ob mice can be largely rescued with exogenous leptin if it is administered when the leptin surge would naturally occur (P4-P14). The temporal window for this effect is tightly restricted (198). Administration of leptin to adult Lepob/ob mice fails to restore hypothalamic projection densities. Early exposure of Lepob/ob mice to exogenous leptin rescues AgRP inputs to preautonomic but not the neuroendocrine neurons within the PVH indicating that, in addition to its role in formation of hypothalamic projections, leptin is also important in the targeting of axons originating from the ARH to specific cell types within the hypothalamus (200).

In mice, the number of Lepr-positive neurons in the ARH increases by roughly a factor of 3 from P5 to P25, at which time adult levels are achieved (201). Lepr is expressed in ∼40% to 50% of NPY- and in less than 20% of POMC-expressing neurons within the first 2 postnatal weeks of life; as the neurons mature, Lepr positivity increases to 80% in these neurons by the third postnatal week. In P13-15 brain slices leptin exposure depolarizes ARH NPY/AgRP/GABA (“NAG”) neurons. By P27, the electrophysiological response to leptin in NAG neurons is mixed with some depolarizing and others hyperpolarizing. Throughout the postnatal, period, hyperpolarizing neurons increase in number. At terminal maturity, by P30, all NAG neurons exhibit the classic inhibitory (hyperpolarization) response to leptin (201). This electrophysiological change in response to leptin in NAG neurons is consistent with leptin's dual roles. During the immediate postnatal period, leptin exposure promotes axonal outgrowth and, after mature feeding circuits are formed, leptin has an inhibitory role to regulate energy homeostasis.

Brain Stem Neurocircuits

Nuclei within the hindbrain—including the nucleus of solitary tract (NTS) and the area postrema (AP)—are involved in feeding behavior but until recently were considered to primarily regulate short-term feeding by responding to visceral signals to control meal termination. However, the NTS also contributes to long-term energy balance. The NTS integrates hormonal and viscerosensory information conveyed through the vagus nerve to coordinate metabolic regulatory processes. A subset of NTS neurons express LepRb; leptin signaling through these neurons affects feeding and energy homeostasis (202). Knockdown of LepRb expression in NTS neurons via AAV-shRNAi injection results in increased food intake and body weight of rats on both chow and HFD (203, 204), while direct leptin injections into the NTS reduce food intake and body weight (205). The NTS is the only site within the CNS that contains neurons expressing Gcg which encodes preproglucagon (206). Preproglucagon is processed into glucagon-like peptide-1 (GLP-1), a peptide that suppresses food intake. These GLP-1 neurons are glutamatergic and project primarily to the PVH. Approximately 60%, 40%, and 26% of the NTS GLP-1-, CCK-, and POMC-expressing neurons, respectively, coexpress LEPR (207), suggesting that leptin signaling may modify the activity of these neurons. In support of leptin importance in GLP-1 and CCK neurons, lack of leptin signaling in LepOb/Ob mice results in increased expression of both of these neuropeptides in the NTS, whereas POMC expression remains unchanged (207). When stimulated chemogenetically using DREADDs, GLP-1/LEPR coexpressing neurons in the NTS only moderately reduce food intake. However, when all NTS LEPR neurons are activated the effect on food intake reduction is significantly stronger—suggesting that NTS LEPR neurons can, independently of GLP-1, suppress food intake (208).

As noted under “Hypothalamic development,” leptin impacts the innervation of hypothalamic neurons from the ARH to other hypothalamic targets during the postnatal period. Biddinger et al showed that axonal contact of GLP-1-expressing neurons in the NTS with the PVH are influenced by leptin as well (209). Projections from the NTS GLP-1/LEPR neurons to the PVH are elevated in Lepob/ob mice at P16, P24, and P60. Restoring leptin signaling to just these neurons (using LepRbTB/TB mice with a transcriptional block flanked by LoxP sites upstream of LepRb which was excised in Cre-expressing GLP-1 neurons (210)) normalized the density of the GLP-1 projections in the PVH, suggesting that leptin reduces targeting of GLP-1 projections to the PVH during development (209). However, this leptin rescue of neuronal projections did not result in any detectable physiological or behavioral feeding improvements in mice fed regular chow. The phenotype was not assessed under conditions of HFD feeding (209). The discovery of increased GLP-1 neurite density from NTS to the PVH in Lepob/ob mice was orthogonal to an earlier-reported decrease in AgRP neurite density from ARH to PVH, DMH, and LHA in the absence of leptin. The former suggests that leptin suppresses axonal outgrowth while the latter indicates a neurotrophic role. In addition to the apparent brain region specificity of leptin's developmental actions, cell specificity was detected. Leptin's effects in the NTS were not uniform on all neuronal inputs into the PVH. Increased GLP-1 inputs onto CRH neurons were noted with no change in the oxytocin neurons (209). This specificity of leptin cellular effects was observed earlier within the hypothalamus: Postnatal leptin supplementation to Lepob/ob mice restored AgRP innervation specifically in the preautonomic neurons of the PVH but not the inputs in the neuroendocrine compartment of PVH (200). It is important to note that leptin exerts diverse and divergent effects on AgRP neurons during both developmental and adult stages. In early development, leptin enhances neurite outgrowth from AgRP neurons while in adulthood, its role shifts to regulating adiposity and inhibiting AgRP neuron activity. In contrast, leptin apparently inhibits GLP-1 axon growth during immediate postnatal period but in adulthood, leptin activates the GLP-1 neurons in the NTS. These data indicate that leptin is necessary for the proper neuro-organization of the hypothalamic and brain stem feeding circuits but its developmental actions are brain region– and cell type–specific, promoting the axonal growth of AgRP neurons and suppressing the GLP-1 axonal outgrowth.

Hedonic Neurocircuits

Leptin mediates the feeding behavior in adult animals in part through its actions on the mesolimbic dopamine (DA) system—one of the critical brain reward pathways originating in the ventral tegmental area (VTA) and projecting to the nucleus accumbens (NAcc), prefrontal cortex, and the amygdala (211). The DA neuron signaling in the VTA to its targets promotes reward-related activities. Food can act as a reward and stimulate the release of DA into the NAcc. Diet-induced obese rats have a reduced basal extracellular DA in the NAcc and release of DA in the NAcc is induced by highly palatable food but not regular chow (212). Human imaging studies suggest that heightened reward responsivity in the NAcc to food cues is associated with indulgence in overeating (213). Functional magnetic resonance imaging in 4- to 6-year-old children found a positive association of eating in the absence of hunger with activity of the NAcc (214).

Dopaminergic and GABA neurons in the VTA express LepRb (215-218) and direct injections of leptin into this area in adult rats cause phosphorylation of STAT3 primarily in DA neurons, a decrease in their firing rate, and a decrease in 24-hour regular chow intake and body weight of the rats (216, 219, 220). Reduction of VTA LepRb with shRNA increases acute HFD food intake and response to progressive ratio for chow (which measures effort expended for food rewards) but had no effect on long-term (4 weeks of HFD feeding) body weight (216, 219). A subset of GABAergic/inhibitory neurons within the LHA also express LepRb and innervate the VTA (221). Direct LHA leptin injection decreases food intake and body weight over a period of 24 hours (221). Decreasing LHA LepRb via direct shRNA injections increased food intake and body weight on HFD over 4 weeks (219). However, since the LHA participates in both homeostatic and hedonic systems the effects of reduced LepRb may be mediated by both. Recently Omrani et al found that leptin activates the LepRb-expressing GABA neurons in the VTA which in turn inhibit DA neurons that project to NAcc to reduce food reward seeking (222) This result suggests that leptin's effects on DA neurons are indirect. They found that optogenetic stimulation of LHA LepR neurons projecting to VTA activates DA neurons via disinhibition of the VTA GABA neurons. Chemogenetic activation of these neurons increases motivation to work for a food reward only in fed (not food-restricted hungry) mice. These results are consistent with leptin's inhibitory effects on VTA-projecting LepRb-expressing LHA neurons (222). In adult animals leptin modulates the DA activity in the reward centers through multiple pathways.

While the development of hypothalamic circuits involved in homeostatic feeding are well described, the data on neurocircuit-formation subserving hedonic systems are limited. Neuronal connections from leptin-responsive neurons in LHA to the VTA were detected within the first week of life in rats (223). The responsiveness of neurons in the VTA and LHA to leptin—defined by activation of pSTAT3 and pERK1/2—is sparse on P10 and more pronounced by P16 (223, 224). Maternal “junk food” diets (consisting of peanut butter, hazelnut spread, cookies, savory snacks, breakfast cereal, processed meat, and a mixture of lard and standard rat chow) fed during lactation are associated with increased preference for fat in the offspring at weaning and at 3 months of age. The offspring of “junk food” fed dams had increased expression of μ-opioid receptor (Mu) and decreased DA active transporter expression in the VTA at 6 weeks of age, but these phenotypes were reversed (decreased Mu and increased DA active transporter expression) by 3 months (225). Another group used RNAseq to compare expression in the VTA and substantia nigra of 6-month-old offspring reared by dams fed HFD during lactation. Expression signatures in the VTA and substantia nigra were altered and the most affected biological functions included neuron development, ion transport, regulation of membrane potential, DA metabolism, locomotion, and behavior. Additionally, the adult offspring of HFD-fed dams had impaired development of the DA neurons and their projections (decreased density of tyrosine hydroxylase) in the substantia nigra and nigrostriatal tract but no changes in the density of DA neuronal fibers in the VTA, no alterations in DA neuronal numbers, and reduced stimulus-evoked DA release in the striatum, a midbrain dopaminergic neuron target site (226). These data suggest that maternal HFD exposure during lactation directly affects the development and function of midbrain dopaminergic circuits in progeny.

We and others have demonstrated that elevating leptin during the first 3 postnatal weeks—a critical time window for brain development—predisposes rodents to subsequent increased weight gain when fed a highly palatable energy-dense diet as adults. Compared with controls, these postnatally hyperleptinemic animals are at the same weight and body composition when maintained on regular chow but gain more weight as soon as they are offered a HFD (more details under “Leptin” within the “Hormones During the Perinatal Period” section below) (149, 227, 228), suggesting that hedonic circuit development (eg, midbrain dopaminergic circuits) may be modulated by early exposure to leptin. More research is warranted to assess the molecular mechanisms that drive changes to the reward system under conditions of maternal HFD feeding during lactation: Does the leptin surge that occurs during this interval influence the development of reward system neurocircuits and subsequent neuronal responsiveness to leptin?

Leptin Effects on Glia

Neurons are not the only cell type affected by leptin during the postnatal brain development. In rodents, during postnatal weeks 2 and 3, glia cell number markedly increases (229, 230), coinciding with the leptin surge (177). The most abundant glial cells in the brain are astrocytes which provide physical neuronal support but also participate in neurogenesis, neuronal development, synaptogenesis, synaptic plasticity, and synaptic transmission (231, 232). Importantly, astrocytes mediate metabolic sensing of nutrients, including glucose (233) and lipid, and energy homeostasis. Astrocytes within the hypothalamus express receptors for and respond to hormones implicated in energy balance, such as leptin (229, 234, 235) ghrelin (236), and insulin (237). Systemic leptin administration between P8-P12 (during the physiological leptin surge) increases proliferation of astrocytes in the hypothalamus; astrocyte-specific deletion of LepRb results in decreased astrogenesis (238). Conditional deletion of LepRb in adult mouse astrocytes leads to morphological changes in astrocytes and increased synaptic inputs onto hypothalamic POMC and AgRP neurons (229). Acknowledging the potential issue of cre drivers being expressed in unintended cell types, these mice display decreased leptin-induced suppression of food intake (229). These data suggest a direct impact of leptin on astrocyte development and function in adult mice.

Early Overnutrition Effects on Feeding Circuits

Offspring of dams fed HFD during gestation and lactation (178) or only during lactation (160) gain more weight than controls during adulthood. The density of AgRP immunoreactive fibers in the PVH is decreased in the offspring of HFD-fed compared with those of chow-fed dams; these progeny display decreased hypothalamic leptin sensitivity (determined by leptin-induced pSTAT3 in the ARH and ventromedial nucleus of the hypothalamus [VMH]) at P30 and P90 (178). Consistent with this finding, Vogt et al reported reduced densities of AgRP and α-MSH (alpha-melanocyte stimulating hormone) fibers from the ARH to functionally downstream hypothalamic areas (PVH, DMH, and LHA) in the offspring of dams fed HFD during lactation [160) (Fig. 3). Maternal HFD feeding increases the projection density of SF-1 and BDNF-positive neurons from the VMH to the LHA (239). Since postnatal overfeeding causes an augmented leptin surge (149), it is surprising that the alterations in hypothalamic circuits reported in postnatally overfed (and therefore postnatally hyperleptinemic) offspring are similar to alterations seen in leptin-deficient mice (198), suggesting that developing hypothalamic feeding circuits are critically sensitive to ambient leptin concentrations. Kirk et al hypothesized that postnatal hyperleptinemia induces leptin resistance thereby attenuating leptin signaling and impairing the development of hypothalamic projections (178). Park et al showed that maternal obesity induces endoplasmic reticulum (ER) stress in tissues that are critical for energy metabolism, including the arcuate nucleus of hypothalamus, during the early postnatal period (Fig. 3). At P14 the POMC and a-MSH axonal densities in the PVH were decreased in pups born to HFD-fed dams compared to control-fed dams, while the densities of AgRP-immunoreactive fibers were not affected. Additionally, leptin signaling (measured by STAT3 phosphorylation) in the offspring of HFD-fed dams was reduced and relieving ER stress in neonatal pups with chemical chaperones such as TUDCA normalized neurostructural changes in the hypothalamus, possibly via improvement in leptin sensitivity (240). Other disturbances to early hypothalamic development induced by maternal obesity include a decrease in proliferation of neural progenitor cells and IR in fetal hypothalamus (241) (Fig. 3). Such fetal IR could be another driver of long-term dysregulation of energy homeostasis in offspring exposed to maternal obesity.

Figure 3.

Figure 3.

Developmental overnutrition effects on feeding circuits. Under conditions of maternal HFD feeding or overfeeding of offspring via reduced litter size during the early postnatal period, the offspring are more susceptible to obesity. This association could be mediated by mechanisms that include an augmented postnatal leptin surge, changes in the quality or quantity of neuronal connections within the feeding circuits, decrease in central leptin and insulin signaling, increase in ER stress within the ARH, decrease length of the hypothalamic primary cilia, alterations in synapses of POMC neurons, and decreased neuronal progenitor cell proliferation. Some or all of these mechanisms are likely affected by early overnutrition and contribute to subsequent susceptibility to obesity. Figure created with BioRender.com.

In addition to quantitative changes of neuronal processes and targets within the feeding circuits, overnutrition during development alters synaptic connections per se (Fig. 3). Two-week-old offspring raised in small litters had an increase in postsynaptic inhibitory currents onto POMC neurons compared with pups from normal-size litters, but by week 3 this phenotype was absent (242). In control mice, leptin effects on the membrane potential of the POMC neurons were heterogenous (stimulatory and inhibitory) in the first 2 weeks of life but transitioned to primarily stimulatory actions in adult mice (242). Leptin did not change inhibitory postsynaptic currents onto POMC neurons from postnatally overfed (via rearing in small litters) mice during the early postnatal period or in adulthood, suggesting that the ability of leptin to reduce the strength of inhibitory inputs onto POMC neurons is blunted by early overnutrition (242) and may be influencing the development of obesity in the offspring.

Maternal HFD feeding during gestation and lactation significantly reduces the length of hypothalamic primary cilia of neonatal pups (243) (Fig. 3). Defects in proteins required for ciliary formation and function cause severe obesity in humans and mice (244, 245). Disruptions in proper development of cilia resulting from maternal nutrition could contribute to the obese phenotype of the offspring. Microglia may be the mediator of hypothalamic feeding circuit programming by maternal nutrition. Microglial activation (measured by morphological changes in Iba1+ cells) was increased in the chow-fed offspring of dams maintained on HFD throughout pregnancy and lactation (246). Depleting microglia from the developing fetuses during gestation by supplementing the diet of pregnant females with colony-stimulating factor 1 receptor inhibitor (PLX5622) causes a reduction in the number of hypothalamic POMC neurons and an increase in offspring weight gain starting on P5 (247).

The primary issue with the maternal HFD model is that it is noisy by virtue of multiple metabolic consequences, complicating identification of primary molecular mechanisms driving the programming of body weight. For that reason, many groups have tested isolated characteristics of the HFD and related obesity models. Concentrations of 4 hormones, leptin, insulin, ghrelin, and amylin, have been shown to be influenced by postnatal overnutrition, and it is plausible that they drive—at least in part—the adult phenotype.

Hormones During the Perinatal Period: Effects on Feeding Circuits and Body Weight in Adulthood

Leptin

Perturbation of circulating leptin concentrations during the postnatal period—specifically, altering the magnitude of the naturally occurring leptin surge between P0 and P22—has long term physiological consequences. Both reduced and augmented leptin surges result in subsequent increased body weight gain when fed high-energy diet or HFD (166, 248). The timing and route of leptin administration has varied in studies of the physiological consequences of postnatal leptin supplementation on adult body weight of the offspring in rodents. Elevating plasma leptin concentration using a TET-On (doxycycline-dependent) leptin transgenic mouse (LepOE) during the period of the natural leptin surge (first 3 weeks of life) increased weight gain when mice were subsequently exposed to HFD at 10 weeks of age (166) (Fig. 4). In this study the LepOE and control pups were littermates, hence prenatal or postnatal environment were the same; the transgenic construct allowed for noninvasive (doxycycline in drinking water) manipulation of circulating leptin in LepOE mice at any point. Importantly, in the same study, exposure of “adolescent” mice (between 3 and 8 weeks old) or adult mice (9-29 weeks old) to doxycycline-induced hyperleptinemia did not cause long-term changes in energy homeostasis of the mice (Fig. 4). The, long-term consequences of transient hyperleptinemia were entirely dependent on the timing of exposure (166). Since overnutrition in mice (either by reducing litter size or maternal HFD feeding) augments the leptin surge, inducing a state of elevated leptin constitutes a physiological and mechanistic surrogate for overnutrition, supporting the inference that the increased adiposity associated with postnatal overnutrition is driven centrally by this augmented postnatal leptin surge. Consistent with this inference, others have reported that intraperitoneal administration of leptin to nursing pups (P0-10, P10-20, or P3-13) increases weight gain in adulthood when the animals are fed a HFD (Table 1) (227, 228).

Figure 4.

Figure 4.

Effects on body weight of transient hyperleptinemia. Transgenic mice that transiently overexpress leptin during the immediate postbirth period have no obvious phenotype until exposed to high-fat chow as adults. This exposure reveals a developmentally programmed susceptibility to increased weight gain on HFD compared with control littermates. When exposure to elevated leptin occurs in adults with a slow and gradual increase in leptin concentrations (to mitigate weight loss), the mice do not show any difference in body weight compared to controls even after HFD challenge. Figure created with BioRender.com.

Table 1.

Effects of hormone exposure during development on the programming of body weight in the offspring

Hormone Effects on feeding circuits and body weight in adulthood
Leptin Augmented postnatal leptin surge → Increased body weight gain in adult mice on HFD (166, 248).
Oral leptin administration during early postnatal development → Protection from DIO in adulthood (249).
Reduced postnatal leptin surge → Increased weight gain in adult mice on HFD (248, 250).
Transient hyperleptinemia effects depend on timing of exposure (166).
Insulin Hypothalamic insulin administration in early postnatal development → Increased body weight gain and glucose impairment (251).
Elevated maternal insulin → Altered hypothalamic circuits and glucose homeostasis (197).
Ghrelin Ghrelin signaling modulation in early postnatal development → Altered hypothalamic neurocircuits, increased body weight and fat (252).
Amylin Amylin administration in early postnatal development to DIO rats → Enhanced leptin signaling, restored hypothalamic neurocircuits; no protection from weight gain in adulthood (253).
Maternal HFD exposure in gestation → Amylin resistance and impaired POMC neuron differentiation (254).
GLP-1 GLP-1R agonist (Exendin-4) administration in early postnatal development to IUGR offspring→ Prevented diabetes, attenuated weight gain in adulthood, and altered neuroarchitecture (255, 256).
Early postnatal Exendin-4 administration in wild type → Reduced body weight and adiposity, and altered neuroarchitecture (256).

Abbreviations: DIO, diet-induced obesity; GLP-1, glucagon-like peptide-1; HFD, high-fat diet; IUGR, intrauterine growth restriction; POMC, proopiomelanocortin.

The concentrations of leptin in breast milk differ between mice and rats, with reported values up to 20 ng/mL in mice (257, 258) and 3 ng/mL in rats (170, 259, 260). Leptin concentration is higher in milk from HFD-fed dams (at P10) than in control diet–fed dams (258, 261). Administration of leptin orally is not equivalent to elevating circulating leptin. Casabiell et al reported that in mice, ingested leptin reaches the circulation (262). Oral administration of leptin from P1 to P20 (estimated to be 5 times the mean daily leptin intake from the mother's milk, by increasing supplementation from 1 to 44 ng per day in each animal over the first 20 postnatal days (263)) resulted in protection from diet-induced obesity (DIO) in adulthood. When postnatally leptin-fed rats (P1-P20) were exposed to HFD from weaning, they had ∼7% lower body weight than controls at 6 months of age (249) and improved insulin and leptin sensitivity (although these results were confounded by significant difference in body weight at the time of measurement (264)). Ingested leptin could drive physiological changes via the intestinal K and/or L cells, which secrete glucose-dependent insulinotropic polypeptide and GLP-1, respectively, which could then influence the sympathetic and/or sensory innervation of the gut resulting in lasting effects on energy homeostasis.

While elevated circulating leptin during the immediate postnatal period is associated with an increase in adult body weight, insufficient postnatal leptin signaling promotes a similar phenotype. Newborn rats administered a leptin receptor antagonist from P2-P13 are more leptin resistant in adulthood—they do not reduce food intake or weight after 1 week of exogenous leptin administration—and gain more weight than controls when fed a high-density diet in adulthood (248, 250). These data suggest that the relationship between circulating leptin concentrations (or the amount of CNS leptin signaling) during the postnatal period and adverse energy homeostatic outcomes in adulthood are U-shaped; too much or too little leptin during the critical developmental time window can predispose to diet-induced obesity in the offspring. The mechanisms by which postnatal leptin is driving this phenotype is not clear but may involve the changes in neuroanatomic architecture of the feeding circuits or alterations to the early neuronal specification that could ultimately result in decreased cellular responsiveness to increasing leptin concentrations. The effects of postnatal leptin elevation could also be conveyed via epigenetic (ie, DNA methylation or histone modifications) changes within the CNS, specifically in the leptin-dependent circuits (more on epigenetics in the following sections).

Insulin

Insulin is implicated as a neurotrophic factor during development. Plagemann et al implanted insulin directly into the hypothalamus of P2 or P8 rats, causing increased body weight gain and impaired glucose homeostasis in adulthood (251). Hypothalamic insulin administration at P8 caused morphological changes within hypothalamic nuclei—including hypotrophy of neuronal nuclei within the VMH and hypertrophy in the DMH—suggesting that postnatal hyperinsulinemia may disturb the neuro-organization of the hypothalamus (265). Subcutaneous injections of long-acting insulin on P8-P11 increased body weight of the rats as early as P10 and throughout lifetime (Table 1). At 7 months of age these rats showed hyperinsulinemia, impaired glucose tolerance, and increased systolic blood pressure; at 30 months of age neuronal number was decreased in the VMH (266). In a study in which wild type offspring born to dams who were either wild type or heterozygous for a null allele of the insulin receptor (hence hyperinsulinemic, insulin resistant), Carmody et al found that maternal IR increased the number of POMC-expressing cells in the ARH of P9 wild-type offspring of the hyperinsulinemic dams (197). These results suggest that maternal insulin influences the specification of POMC neurons (197). Vogt et al showed that elevated maternal insulin resulting from maternal HFD exposure during lactation is associated with decreased α-MSH and AgRP fiber density in the PVH, DMH, and LHA in 8-week-old offspring. They then blocked insulin signaling specifically in POMC neurons of the offspring only (not the dams, using insulin receptor flox/flox [IRfl/fl] dam crossed with IRfl/fl and POMC-Cre sire), under conditions of maternal HFD feeding (causing maternal hyperinsulinemia), and found that lacking insulin signaling in POMC neurons rescues the projections in this circuit and corrects glucose intolerance but does not prevent increased body weight (160). These results implicate a role of POMC insulin signaling in the development of hypothalamic circuits involved in glucose homeostasis.

Ghrelin

Ghrelin—an appetite-stimulating circulating factor—is primarily produced in the stomach and signals through the growth hormone secretagogue receptors located across the CNS (267-269). Neural expression for growth hormone secretagogue receptors is highest in the hypothalamus (including ARH, VMH, and lateral mamillary nuclei) (269); administration of ghrelin activates neurons in brain areas known to control feeding (such as ARH, VMH, and PVH) (270). In wild-type mice circulating ghrelin levels rise from P6 and reach adult concentrations by P14 (252). To evaluate the physiological and neurobiological roles of ghrelin during postnatal development, ghrelin signaling was either blocked (by daily intraperitoneal injections of an antighrelin compound that specifically binds and inhibits the bioactive acylated form of ghrelin) from P4 to P22 or circulating ghrelin concentrations were elevated (via daily intraperitoneal injections resulting in at least 50-fold increase in plasma concentration; ghrelin concentrations were increased within 15 minutes of injection and returned to control concentration within 12 hours) from P4 to P12. Blocking ghrelin action enhanced overall PVH fiber density, including AgRP and αMSH immunoreactive fibers; administration of ghrelin had the opposite effect—PVH innervation was reduced. Surprisingly, both interventions resulted in increased body weight, adiposity, and circulating blood glucose. While the overall density of AgRP and αMSH fibers in the PVH are oppositely affected by ghrelin and antighrelin administration, the ratio of orexigenic AgRP to anorexigenic αMSH fibers is increased in both conditions potentially explaining the increased body weight seen with both manipulations (271). These data emphasize that both the developmental timing and the magnitude of ghrelin signaling are important in the proper formation of hypothalamic neurocircuits and are critical in the control of body weight later in adulthood (Table 1) (252). Postnatal overnutrition induces central ghrelin resistance (demonstrated by an attenuated induction of c-Fos immunoreactivity in the ARH following peripheral ghrelin injection) and supports the evidence that hypothalamic development is influenced by ghrelin during the postnatal period (272).

Amylin

Amylin, or islet amyloid polypeptide, is coreleased with insulin from the pancreatic beta cells (273) in molar ratio of 1: up to 100 (274-276). Amylin complements insulin's actions to regulate postprandial glycemia via suppression of glucagon secretion (277). Amylin inhibits food intake and delays gastric emptying via heterodimeric receptors (278) composed of the calcitonin receptor (CTR a or b isoform) and 1 of the 3 receptor activity modifying proteins (RAMP 1-3) in the AP which project to the NTS and lateral parabrachial nucleus. In the hypothalamus, amylin enhances leptin signaling in the VMH and ARH by elevating microglial interleukin-6 secretion which amplifies pSTAT3 expression (253, 279-281).

During gestation, amylin is initially detected in pancreatic islet β-cells at E12 and peaks at E17 (282, 283); the amylin receptor, CTR, is expressed in the brain starting at E12-13. Amylin knockout mice have fewer microglial cells in the ARH and AP at E12; neuronal numbers are unchanged (284). Postnatal neurotrophic effects of amylin have also been reported using RAMP 1/3 knockout mice and knock down of CTR in brains of P4 rats (both of which result in reduced amylin receptor signaling) and amylin KO mice, all showing reduced α-MSH fiber projections from ARH to PVH. The density of AgRP/NPY fibers was higher in amylin KO mice, lower in RAMP 1/3 KO mice and not detectably different in the rats with CTR knocked down specifically in the VMH at P4 (281). Amylin induces p-ERK specifically in arcuate POMC neurons but not AgRP/NPY neurons, suggesting that the effects of amylin to enhance outgrowth of POMC neurons may be direct while those on AgRP/NPY may be indirectly mediated (eg, via a microglia-secreted interleukin-6–related mechanism) (281). DIO rats selectively bred to gain excess weight on high-energy diet are obese and leptin resistant (reduced pSTAT3 activation in the ARH following leptin administration) in adulthood when fed HFD. At P12 these animals have decreased fiber densities from ARH to the PVH (by 1,1′-Dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate (DiI), a fluorescent lipophilic tracer that labels axonal projections labeling) (285). Administering amylin to these DIO rats during the postnatal period (P0-P16) restores a-MSH and AgRP/NPY projection densities in the PVH and improves leptin signaling; however, amylin administration does not protect these animals from excessive weight gain in adulthood (253). Maternal HFD during gestation leads to increased serum amylin and reduced hypothalamic Ramp3 mRNA and protein in male fetuses at E16.5 (254). Decreased hypothalamic Ramp3 causes amylin resistance (decreased amylin-induced phosphorylation of STAT3 and ERK used as surrogates) which impairs differentiation of POMC (but not AgRP/NPY) neurons in hypothalamic neural stem cells (254). Gestational elevation of fetal serum amylin via subcutaneous injections into pregnant dam resulted in decreased body weight and increased hypothalamic Pomc expression and protein level in male offspring compared with controls. These data suggest that amylin plays an important role in differentiation of POMC neurons. Amylin influences the establishment of neurocircuits within, not only the hypothalamus, but also the AP projections to the NTS within the brainstem. Neurite outgrowth (measured with DiI inserted in the AP) from AP to the NTS in amylin KO mice at P10 and P60 showed reductions relative to wildtype mice in neuronal fiber densities from AP to the NTS (286) confirming amylin's role in development of brainstem circuits (Table 1). Similar to insulin and leptin, in human placenta perfused with pramlintide (an analog of human amylin) ex vivo, only a negligible amount of pramlintide crossed the placenta from maternal to fetal circulation (287). Therefore, circulating maternal amylin is unlikely to contribute meaningfully to the increased fetal serum concentration under maternal HFD feeding conditions (254). Possibly, maternal obesity or nutritional status (such as specific macronutrients, hormones, inflammatory molecules or other factors) may directly induce placental or fetal amylin expression. Subcutaneous amylin injections to pregnant mouse dams caused a 2-fold amylin increase in E16.5 fetal serum, suggesting that maternal amylin induces an increase in fetal amylin concentrations (254).

GLP-1

GLP-1, expressed in the intestinal L-cells and NTS neurons, acts in the periphery to slow gastric emptying as well as centrally to reduce food intake and stimulate glucose-induced insulin secretion (288). In a model of IUGR induced by uterine artery ligation in pregnant rats, the offspring have low birth weight and develop obesity and diabetes in adulthood (255, 289). Subcutaneous administration of Exendin-4—a GLP1R agonist—during early postnatal period (P1-P6) to these IUGR offspring prevents the development of diabetes and attenuates weight gain in the adulthood (255). Interestingly, reduced weight gain was also observed in control animals given Exendin-4 on P1-6 (255, 256). Neonatal (P1-P6) administration of Exendin-4 to wild type mice resulted in lower body weight (as early as at P28) and adiposity when maintained on low fat chow (Table 1). Females (but not males) gained less weight when fed 45% HFD, driven by increased energy expenditure, compared with controls. In addition to physiological changes, neonatal Exendin-4 administration reduced NPY neuronal fiber density in the PVH of 4-week-old female mice, an effect that was reversed in animals with Glp1r knocked out in PVH Sim1 neurons (256). This study suggests that manipulation of GLP-1 concentrations early in the postnatal period may affect the neuroarchitecture of the hypothalamus with physiological consequences on body weight in adulthood (in a sex-specific manner) and specifically implicates the GLP1R signaling pathway within the PVH (Table 1).

In addition to the hormones described above, several others, including glucagon, glucose-dependent insulinotropic peptide, and peptide YY, are recognized as important players in regulating energy homeostasis. These hormones may contribute to the developmental programming of adiposity, although direct investigations in this regard are currently lacking. Further research is needed to elucidate their specific roles in shaping adiposity and overall metabolic health during development.

Epigenetic Changes

Epigenetic changes during gestation or the postnatal period are another potential mechanism mediating developmental programming of body weight (290). Epigenetic modifications can alter gene expression without changing the DNA sequence; instead, expression is altered by DNA methylation, histone modifications, and microRNAs (miRNAs). Patterns of DNA methylation have been studied in the context of developmental programming of weight in humans and rodents. Alterations of DNA methylation incurred during development can persist into adulthood, making such alterations a plausible mechanism for developmental programming of body weight by virtue of their “memory” of the nutritional, metabolic and other environmental characteristics. DNA methylation patterns of genes critical in energy and glucose homeostasis, metabolism, and insulin signaling can be altered by exposure of progeny to maternal obesity, HFD-feeding, or overnutrition during gestation and/or lactation (291-297). Overnutrition of weanling rats (by decreasing litter size) during the postnatal period results in hypermethylation of the hypothalamic POMC promoter (293) which is negatively correlated with hypothalamic POMC expression adjusted for circulating leptin. Similarly, offspring of dams fed HFD during pregnancy and lactation show increased methylation of the POMC promoter even when fed regular chow after weaning (291, 298). Maternal HFD feeding results in hypermethylation of the ARH InsR promoter and an associated decrease in InsR expression in the ARH of male offspring only (299). It is unclear by what molecular physiological mechanisms gestational and early postnatal states are conveyed directly to the genome.

Investigating the effects of perinatal nutrition on epigenetic changes in humans is challenging since the tissues of interest (ie, specific brain regions) cannot be directly assessed. As a proxy for overall methylation status some studies assess methylation in cord blood DNA. Even though a number of studies report associations of DNA methylation patterns in cord or newborn blood with maternal BMI or gestational weight gain, the individual genes identified are not consistent across the studies (300-303). In a meta-analysis (integrating epigenome-wide association studies from 19 cohorts), prepregnancy BMI was associated with small variations in DNA methylation of newborn blood at 86 CpGs sites; the effect size for each of the sites was less than a 0.15% change in methylation per unit of maternal BMI increase (304). Following further analyses with causal inference strategies, a support for causal intrauterine effect was found in only 8 out of 86 CpG sites (304). A major limitation of studies associating DNA methylation with maternal BMI or other variables is that they use circulating blood cells as a surrogate for specialized cells in brain, adipose tissue, liver, and other organs. Another meta-analysis of epigenome-wide association studies found that there are only minimal associations between the DNA methylation of specific CpG sites in cord or whole blood from children and adolescents and the BMI in children 2-18 years of age (305). While the authors conclude that differences in DNA methylation are likely a consequence rather than a cause of obesity, it is equally likely that the methylation states of cord or peripheral blood does not reflect the patterns of methylation in tissues that are more relevant to energy homeostasis. One study compared the whole blood methylome of siblings born before and after maternal bariatric surgery to investigate the effects of maternal weight loss and found changes in methylation of genes involved in leptin and insulin receptor signaling, and genes associated with type 2 diabetes (306).

Effects of maternal obesity on placental DNA methylation could more directly reflect intrauterine environment and placental function. DNA methylation in the placenta is increased overall by 21% in obese compared with lean gravidas (307). In genome-scale DNA methylation analysis, altered placental methylation of 56 CpGs was associated with maternal BMI. The LEP promoter is hypermethylated, and the adiponectin promoter hypomethylated on fetal and maternal sides of the placenta, respectively, in obese compared with lean gravidas (308, 309). Similar to maternal obesity effects, Lesseur et al reported increased placental (average of maternal and fetal sides) leptin promoter methylation in infants exposed to GDM (308). In contrast, Bouchard et al observed a negative and positive correlation between glucose levels (at 24-28 weeks) and LEP methylation on the fetal and maternal side, respectively (310). Since hypermethylation is associated with reduced gene expression, maternal obesity and diabetes may influence leptin production by the placenta, and the intrauterine milieu affecting fetal development. This is a complicated interplay since it is also possible that leptin (or other metabolites/ hormones) during development influence the patterns of DNA methylation.

Animal studies on ancestral exposure to HFD, malnutrition, and environmental toxicants have demonstrated that obesity risk can be conveyed transgenerationally through epigenetic modifications (311). One study found that maternal HFD exposure results in increased body mass in third-generation (F3) female (only) offspring that is transmitted only via the paternal lineage (312). A recent study found that maintaining male mice on a Western diet (high in fat and sugar) for 5 consecutive generations resulted in a progressively amplified obesity phenotype and related metabolic diseases in the offspring (313). Furthermore, even when the progeny of these multigenerational Western diet–fed males were fed a standard diet, both males and females remained heavier than controls for 4 subsequent generations (313). This transgenerational obesity may be mediated by epigenetic marks. The Agouti viable yellow (Avy) mouse has been investigated as a model of maternal obesity. The Avy is a metastable epiallele—an allele of the same gene that differs in DNA methylation status and is susceptible to methylation changes by environmental factors—allowing for isogenic mice to exhibit different body weight and fur color phenotypes (314). Undermethylation of the Avy allele results in increased expression of ASP (which blocks MSH activity at melanocortin receptors) and obese mice with yellow coats (Avy/a), while mice with a silenced allele are lean with agouti fur. In successive generations of obese Avy/a female mice Waterland et al found that with every successive generation maternal obesity increased and body weight in adult progeny increased incrementally (by generation) while maintained on standard chow (315). A contemporaneous cohort of mice fed a methyl-group supplemented diet (supplemental folic acid, vitamin B12, betaine, and choline), inducing developmental DNA hypermethylation, did not experience the same incremental increase in offspring obesity with subsequent generations. The coat color phenotype—used as a proxy for Avy methylation—was not associated with body weight in either group, suggesting that maternal obesity affects the developmental establishment of epigenetic modifications at other genomic loci critical in body weight regulation (315). Consistent with the idea that obesity inhibits methylation and this reduced methylation status confers offspring obesity risk, supplementation of methyl donors to HFD-fed C57BL/6J dams during gestation and lactation mitigates the increase in body weight found in adult offspring born to dams fed HFD (316). Promoter region methylation of obesity-related genes, Pparg, Fas, Lep, and Adipoq was reduced in the adipose tissue of offspring reared by HFD-fed mothers but was mostly rescued by dietary supplementation of methyl donors in the HFD. With the exception of Lep, gene expression was inversely related to methylation status of these genes (316). Another group carried out a similar experiment and found that maternal consumption of HFD during pregnancy and lactation results in global DNA hypomethylation in the offspring brain and this effect can be reversed by supplementing HFD with methyl donors (317, 318). Body weight gain in the offspring of HFD-fed mothers was attenuated with dietary methyl group supplementation at 12 weeks of age, however this effect was transient as no body weight difference was detected among the groups (including offspring born to chow vs HFD-fed dams) at 20 weeks of age and beyond. These data support the role of epigenetic modifications in vertical transmission of obesity. The role—if any—of maternal folate status on progeny birth weight in humans is unclear.

It is important to point out that the epigenetic data supporting transgenerational effects of HFD are not consistent across the various studies which may be partially attributed to heterogeneity in epigenetic status by cell type and by some studies being underpowered. These differences underscore the need for further research with improved study designs as suggested by others (319, 320). Such improvements would include interrogating epigenetic status of single cells, having a detailed a priori analysis plan, and validation of results in independent experiments.

Relevance to Humans

Comparison of Brain Development in Human vs Rodent

The timing of brain development between humans and rodents differs considerably; but the sequence of events occurring during brain maturation is largely the same (321) (Fig. 2). Rodents at parturition have less mature brains than humans at birth and experience an accelerated postnatal development (321). Neurogenesis in rodents starts at E9.5 days and lasts through the second postnatal week, while in humans it is estimated to begin at E35 weeks and lasts generally until midgestation but may continue through gestation and postnatally to a limited degree in certain regions such as hippocampus (185). Neurogenesis has been reported in dentate gyrus of adult rodents (322, 323) and humans (324). Neurogenesis specifically in the hypothalamus of mice takes place between E10 and 16 (188, 325) while in humans between gestational weeks 7 and 10 (326), but may also continue in the second trimester (327). Synaptogenesis in humans starts around week 20 of gestation, peaks around 2 years of age, and persists until adolescence. In rodents, synaptogenesis starts at E16 and is completed roughly by weaning but hypothalamic synaptogenesis during which the feeding circuitry matures occurs postnatally, between approximately P6 and 16 (199). The process of myelination also varies among brain regions and species. In humans, myelination begins in the third trimester of gestation and continues postnatally throughout childhood and adolescence and into adulthood (328, 329). In rodents, myelination begins after birth and continues until 6-7 weeks of age (330, 331). Myelination of the hypothalamus in rodents occurs around week 4 and 5 of life (330). Brain growth rate (defined by mass) peaks at the end of third trimester (∼36-40 weeks of gestation) in humans and at P7-P10 in rodents (332-334). The third trimester of human pregnancy is analogous to P1-P10 in rodents; the human neonate's brain development, by mapping cellular transcriptional profiles, corresponds to that of a P7-P10 mouse (186). Based on these estimates, experiments investigating the nutritional environment during early postnatal brain development in mice are relevant to late gestation in humans.

Adiposity in Fetus and Neonates

In the human fetus, accrual of body fat begins at ∼25 weeks of gestation. Based on body composition estimates measured with air displacement plethysmography (ADP, peapod) in healthy preterm infants, the mean fat mass at 30 weeks of gestation is 0.1 kg (7.4% total body mass) and increases to 0.28 kg (10.1%) by 36 weeks (335). These estimates are compatible with fetal chemical analyses (336). In normal term infants, fat mass increases to 0.4 kg (11.2%) by 40-41 weeks of gestation (ADP) (337) and to ∼2 kg (∼26%) by 6 months postnatal (ADP). By 12 months of age, fat mass ranges from 23% to 28% of body mass, and by 3-5 years of age, fat mass percentage drops and ranges from 14% to 24% (depending on gender and ethnicity (338);).

Leptin in Pregnancy and in the Human Fetus

In adult humans, leptin is secreted from adipose tissue in proportion to adipose tissue mass and fat cell volume. During pregnancy, the placenta is an additional source of leptin (339, 340). In nonhuman primates (341) and humans (342-345), maternal circulating leptin increases throughout pregnancy. This increase occurs even during the first trimester, prior to any substantial maternal fat mass increase (344), suggesting that the placenta may be a contributor to the maternal circulating leptin. Most studies report a 1.5- to 3-fold increase in circulating leptin in the second and third trimesters (342-346) with a significant decline in the early postpartum period, as early as 24 hours after delivery (347, 348). Since circulating leptin concentrations are proportional to fat mass prior to pregnancy, overweight or obese pregnant women have higher circulating leptin at baseline. However, the rate at which circulating leptin increases during gestation is significantly lower in overweight/obese than in normal-weight gravida (349). Circulating leptin increases as little as 10% from the first to third trimester in overweight/obese pregnant women compared with a 54% increase in normal weight gravida [349-351). In lean gravidas, Misra et al observed a significant increase in plasma leptin concentrations per unit of body weight with pregnancy progression while the trend was opposite in overweight/obese group suggesting a lower production of leptin (either per fat or placental mass) in overweight/obese gravida in advanced pregnancy (350).

While leptin is detectable by 19 weeks of gestation in human fetal circulation, it is unclear which tissue is its primary source. Fetal plasma leptin concentration correlates with gestational age, fetal size, and birthweight (352). All 45 studies included in a meta-analysis reported a positive correlation between plasma leptin concentration and birth weight (353). Fetal plasma leptin correlates with fat mass, and the slope of the linear regression of gestational week vs circulating leptin during weeks 21 to 32 of gestation is low but increases between weeks 33-41. This rapid increase in slope is associated with a more rapid increase in fetal fat mass (342). Infants born prior to 30 weeks of gestation had ∼6-fold lower circulating leptin concentration than those born between 30 and 36 weeks of gestation (354). Compared to appropriate for gestational age (AGA) controls, IUGR fetuses have lower circulating leptin concentrations (355, 356) likely due to decreased adiposity since leptin per kilogram of mass is unchanged. Fetuses from gestational diabetic (GDM) gravidas have higher leptin than AGA fetuses, even when leptin is corrected for fetal weight (352, 356). Infants born to women with GDM have increased fat mass compared with infants of mothers with normal glucose tolerance (71, 72, 74), suggesting that elevated circulating leptin in these infants is likely due to higher fat mass. Cord blood leptin is also significantly higher in the offspring of women with T1D, and is likely mediated via an increase in fat mass [75). As mentioned under “Perinatal programming of body weight: evidence from humans,” infants born to diabetic mothers have elevated insulin concentration which are likely driving the increase in fat mass accrual during gestation, ultimately resulting in elevated fetal plasma leptin concentrations.

Also, LGA babies have higher, while SGA babies have lower circulating leptin concentrations (357, 358) than AGA, with concentrations that are directly proportional to fat mass (359). This dependence of plasma leptin on fetal size suggests that fetal adipose tissue is an important leptin source in fetal circulation. This interpretation is somewhat confounded by the fact that mothers with GDM are often obese and therefore have higher plasma leptin concentrations; in addition, placental weights are higher in GDM pregnancies (360). However, as mentioned above, infants of gravida with diabetes accrue more fat and hence more leptin. There are also multiple reports of leptin concentrations being significantly higher in female vs male newborns (355, 361-364). Based on body composition of the reference infants, there is no significant difference in fat mass between male and female at birth (365, 366), suggesting that this sexual dimorphism is independent of fat mass. There are mixed reports regarding differences in plasma leptin concentrations in male vs female prepubertal children, but the data are consistent in that circulating leptin is higher in females than males starting from late puberty (367-372) and persists into adulthood even when adjusted for fat mass (373-375). The production of leptin is higher in subcutaneous than in visceral adipocytes. As females tend to have more subcutaneous fat than males, this difference in leptin production accounts in part for the higher concentrations of circulating leptin (adjusted for body fat) in females than males (373). However, it is not entirely clear at which point during development this difference in leptin production emerges.

To determine the extent of placental syncytiotrophoblast contribution to circulating leptin in the human fetus, 3 groups assessed leptin secretion in human ex-vivo placentas (376-378); 2 groups found that only a small fraction (1-5%) of placental leptin is secreted into the fetal side of the placenta with the remainder secreted into maternal circulation, suggesting that the placenta only supplies limited amounts of leptin to the fetus. However, 1 study showed that 46% of placental leptin is secreted to the fetus (378). Since maternal plasma leptin concentrations are ∼2 to 3 times higher than fetal and the fetal blood volume is much smaller than maternal; even ∼5% of placental leptin secreted to the fetal side could be a substantial contributor to circulating fetal leptin. Additionally, as gestation progresses, the weight of the placenta increases. The weight of the placenta between 23 and 27 weeks of gestation increases by 44 g (379) and those delivered between 37 and 41 weeks increases by 59 g (380), indicating that the placenta—whose weight gain parallels the increase in fetal mass and fetal plasma concentrations—could be an additional source of leptin. Most clinical studies do not find any correlation between maternal and fetal/cord plasma leptin concentrations (multiple references (381)). In a study of healthy pregnant women, leptin concentrations in infants classified as small, appropriate, or LGA correlated with their ponderal indices (kg/length m3) (358) but were not related to maternal or placental leptin. Cord blood leptin concentration correlated with fetal size and fat mass suggesting again that fetal adipose tissue may be a more important source of circulating fetal leptin.

As in humans, circulating leptin concentration increases during mouse pregnancy but to a much greater degree (20- to 40-fold). In contrast to humans, the maternal hyperleptinemia in mice is primarily a result of increased placental production of soluble leptin receptor (LepRe isoform) which binds leptin in circulation (382). Similar to humans, secretion of leptin from placenta into fetal circulation in rodents is low (383). 125I-leptin is transported from maternal circulation to the placenta and fetus. This transplacental leptin transfer increases by 10-fold between day 16 and day 22 of gestation in rats (383). The magnitude of molecular leptin transfer from maternal to fetal circulation is unclear and was not addressed by this study. In a study of pregnant leptin deficient (Lepob/ob) females carrying ob/+ fetuses, no detectable leptin was found in maternal circulation, suggesting that leptin does not cross back into maternal circulation from the fetus (382, 384).

Plasma leptin concentrations in nonhuman primates during the third trimester of gestation were 46-fold higher in the maternal vs fetal circulation under regular diet conditions but in mothers fed a 30% HFD, leptin in maternal circulation was doubled while the fetal leptin remained the same. The low fetal compared with maternal leptin concentrations and the lack of increase in fetuses of HFD- vs regular diet-fed mothers suggest that little or no maternal leptin is transported across the placenta (385).

Leptin concentrations in pregnant women increase throughout gestation and drop significantly shortly after parturition, before maternal body fat changes significantly (344). Leptin concentrations in the progeny decline immediately after birth relative to cord blood concentrations. Schubring et al found that leptin in venous cord blood at term of neonates was 3.47 ng/mL, dropped to 0.26 ng/mL by 56 to 79 hours after birth, and remained low (0.59 ng/mL) at 99 to 128 hours after birth (386). Hytinantti et al reported a decrease in cord blood leptin from 9.7 ng/mL at birth to 2 ng/mL by postnatal day 3 (387). Ertl et al reported cord blood leptin at birth of 4.5 ng/mL, decreasing to 0.6 ng/mL by P7 (361). Many other studies have reported 4 times higher values at birth compared with plasma leptin concentrations within a few days after birth (357). Even though there is a decline at birth, leptin concentrations after birth still correlate with body mass. As discussed earlier, leptin concentrations in human fetuses increase during gestation peaking near the end of gestation. Similar to rodents—which have a leptin surge in the immediate postnatal period when the hypothalamic circuits are being formed—the timing of this “leptin surge” in humans is contemporaneous with the development and maturation of the feeding neurocircuits within the hypothalamus.

Rodents are not the only species that experience a postnatal leptin surge. Lambs increase leptin concentration in the early postnatal period. Plasma leptin increases from 1 to 4 ng/mL in lambs between P6 and P10 (388-390) born to ewes fed standard chow. In contrast to rodents, gestational undernutrition or overnutrition eliminates the leptin surge in lambs (at least in the first 10 postnatal days, when the measurements were made); both nutritional manipulations lead to increased food intake and adiposity in the offspring compared with controls (391, 392). In postnatal cows, plasma leptin increases from birth to P2 and then decreases until P16 of age. One study reported stable circulating leptin concentrations in P3 to P8 postnatal pigs while another group found that plasma leptin during the first measurement at P5 was higher than that at P9, 16 or 23. In the aggregate, there is some evidence supporting the presence of postnatal leptin surge in neonatal lambs, cows, and pigs, but it is unclear whether the equivalent may be occurring in utero in nonhuman primates. While longitudinal leptin concentrations in fetal and neonatal nonhuman primates have not been reported to an extent that would allow identification of a postnatal leptin surge per se, similar to humans, plasma concentrations of leptin increase in fetuses of these animals with the progression of gestation (393). As in humans, maternal plasma leptin concentrations during gestation increase in nonhuman primates with the progression of pregnancy and decline after parturition. The magnitude of the pregnancy-associated leptin increase in nonhuman primates is higher than in humans in both absolute concentration and relative to nonpregnant state.

The precipitous decrease in leptin at birth may function as a signal to promote feeding in the neonate. This hypothesis is consistent with the observation that in mice during the postnatal leptin surge, leptin does not elicit anorexic response; mice do not decrease food intake in response to exogenous leptin prior to ∼P15 (166). Immediately after leptin surge (coincidental with maturing of neuronal feeding circuits), mice start to respond to leptin by reducing energy intake. As discussed above, leptin has a dual role as a neurotrophic factor and a regulator of energy homeostasis; the leptin surge may be the inflection point between the 2 functions; until the leptin surge (including the period of elevated leptin), leptin plays a neurotrophic role and, immediately post leptin surge, after plasma leptin concentrations drop to the levels proportional to fat mass, leptin switches primarily to its role as fat mass regulator.

In humans, circulating fetal leptin concentration correlates with birth weight but not with maternal circulating leptin concentrations, and maternal circulating leptin concentrations peak midgestation decreasing towards term, while fetal leptin is highest at the end of gestation. This divergence, coupled with the fact that leptin concentrations decrease dramatically after birth, suggests a contribution of placental leptin production to fetal circulating leptin. It is also possible that there are in utero signals stimulating fetal leptin secretion either from fat or placental tissue and that this stimulation ceases abruptly at birth. Leptin acts as a predictive early starvation warning signal, dropping rapidly at the onset of negative energy balance in adults. The decline in leptin at birth could act as an anticipatory starvation signal for the weight loss that occurs within the first week after birth.

Important Considerations

Critical maternal hormones, including insulin and leptin, do not cross the placenta to any significant degree, therefore, direct effects of these hormones on the developing fetus are unlikely. One possibility is that developmental programming is mediated by increased placental insulin and leptin signaling, inducing epigenetic changes to the placenta directly altering placental function. Potential functional changes include transferring nutrients across the placenta, increasing placental oxidative stress and inflammation. Under maternal hyperglycemic conditions, increased glucose transfer from maternal to fetal circulation leads to fetal hyperinsulinemia which contributes to increased fat accrual and this increased adiposity results in elevated fetal leptin concentrations. It is possible that elevated endogenous fetal insulin and leptin are the primary effectors causing altered CNS development. Alternatively, maternal circulating hormones such as insulin and leptin may be acting as biomarkers for some other unknown factors that do cross the placenta and confer effects on the hypothalamic development and ultimate programming of adiposity.

Understanding the neuromolecular pathways for the developmental programming of adiposity maybe be helpful in better informing pregnant women about gestational obesity and diabetes consequences as well as for the discovery of new therapeutics to treat transgenerational obesity. Numerous studies indicate that pregnancy is the 1 time during which people are more motivated to modify difficult behavior for the betterment of their offspring. Current interventions are limited primarily to lifestyle changes (dietary and exercise) during pregnancy. However, these interventions are minimally effective at improving progeny outcomes such as decreasing the risk of high birth weight (394) and decreasing the rate of fetal adipose deposition during pregnancy (395). Other studies have not found improvement in the infant birth weight following maternal lifestyle interventions (396). Reducing body weight and improving metabolic health in women prior to conception could have the most profound impact, not only on the maternal and progeny outcomes, but also on the health of future generations.

Conclusions

In this review we discuss neuromolecular mechanisms by which aspects of nutritional status during early life can induce persistent changes within the CNS which influence subsequent weight homeostasis. Developmental overnutrition in rodents via maternal HFD feeding or postnatal overfeeding by litter size reduction leads to obesity in adulthood. The mechanisms by which this programming of body weight occurs are complex. The data currently available suggest that attention to quantitative and qualitative aspects of maternal body mass/composition and nutrition during the third trimester and to neonatal nutrition could impact long-term susceptibility to obesity by effects on the fine structure of specific regions of the brain. Large clinical studies to define and quantify these factors may not be feasible or worthwhile, but animal models could point to specific mechanisms that could inform focused hypotheses that would be testable in limited numbers of human subjects.

Abbreviations

ADP

air displacement plethysmography

AGA

appropriate for gestational age

AgRP

agouti-related protein

AP

area postrema

ARH

arcuate nucleus of the hypothalamus

BMI

body mass index

CNS

central nervous system

DA

dopamine

DIO

diet-induced obesity

GCK

glucokinase

GDM

gestational diabetes

GLP-1

glucagon-like peptide-1

HFD

high-fat diet

InsR

insulin receptor

IR

insulin resistance

IUGR

intrauterine growth restriction

Lepr

leptin receptor

LGA

large for gestational age

MODY

maturity-onset diabetes of the young

NAcc

nucleus accumbens

NPY

neuropeptide Y

NTS

nucleus of solitary tract

POMC

proopiomelanocortin

SGA

small for gestational age

T1D

type 1 diabetes

T2D

type 2 diabetes

VTA

ventral tegmental area

Contributor Information

Alicja A Skowronski, Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA.

Rudolph L Leibel, Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA.

Charles A LeDuc, Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA.

Funding

This work was supported by research grants from the National Institutes of Health: R01 DK052431 to R.L.L., and the NY Nutrition and Obesity Research Center: P30 DK026687 to R.L.L.; and Pfizer in the form of an Pfizer Obesity ASPIRE grant to C.A.L.

Disclosures

The authors declare no competing financial interests.

References

  • 1. Fryar CD, Carroll MD, Ogden CL. Prevalence of Overweight, Obesity, and Severe Obesity among Children and Adolescents Aged 2–19 Years: United States, 1963–1965 Through 2015–2016. National Center for Health Statistics (U.S.); 2018. [Google Scholar]
  • 2. Must A, Must A, Jacques PF, et al. Long-term morbidity and mortality of overweight adolescents. A follow-up of the harvard growth study of 1922 to 1935. N Engl J Med. 1992;327(19):1350‐1355. [DOI] [PubMed] [Google Scholar]
  • 3. Park MH, Falconer C, Viner RM, et al. The impact of childhood obesity on morbidity and mortality in adulthood: a systematic review. Obes Rev. 2012;13(11):985‐1000. [DOI] [PubMed] [Google Scholar]
  • 4. Abdullah A, Wolfe R, Stoelwinder JU, et al. The number of years lived with obesity and the risk of all-cause and cause-specific mortality. Int J Epidemiol. 2011;40(4):985‐996. [DOI] [PubMed] [Google Scholar]
  • 5. Demeulemeester F, de Punder K, van Heijningen M, et al. Obesity as a risk factor for severe COVID-19 and complications: A review. Cells. 2021;10(4):933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Pellegrini M, Ponzo V, Rosato R, et al. Changes in weight and nutritional habits in adults with obesity during the “lockdown” period caused by the COVID-19 virus emergency. Nutrients. 2020;12(7):2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Deschasaux-Tanguy M, Druesne-Pecollo N, Esseddik Y, et al. Diet and physical activity during the coronavirus disease 2019 (COVID-19) lockdown (march–may 2020): results from the French NutriNet-santé cohort study. Am J Clin Nutr. 2021;113(4):924‐938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. He M, Xian Y, Lv X, He J, Ren Y. Changes in Body Weight, Physical Activity, and Lifestyle During the Semi-lockdown Period After the Outbreak of COVID-19 in China: An Online Survey. Disaster Med Public Health Prep. 2021;15(2):e23‐e28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Vogel M, Geserick M, Gausche R, et al. Age- and weight group-specific weight gain patterns in children and adolescents during the 15 years before and during the COVID-19 pandemic. Int J Obes. 2022;46(1):144‐152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Stavridou A, Kapsali E, Panagouli E, et al. Obesity in children and adolescents during COVID-19 pandemic. Children (Basel). 2021;8(2):135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Dunton GF, Do B, Wang SD. Early effects of the COVID-19 pandemic on physical activity and sedentary behavior in children living in the U.S. BMC Public Health. 2020;20(1):1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Xiang M, Zhang Z, Kuwahara K. Impact of COVID-19 pandemic on children and adolescents’ lifestyle behavior larger than expected. Prog Cardiovasc Dis. 2020;63(4):531‐532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Prospective Studies C. Body-mass index and cause-specific mortality in 900 000 adults: collaborative analyses of 57 prospective studies. Lancet. 2009;373(9669):1083‐1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Fontaine KR, Redden DT, Wang C, et al. Years of life lost due to obesity. JAMA. 2003;289(2):187‐193. [DOI] [PubMed] [Google Scholar]
  • 15. Lung T, Jan S, Tan EJ, et al. Impact of overweight, obesity and severe obesity on life expectancy of Australian adults. Int J Obes (Lond). 2019;43(4):782‐789. [DOI] [PubMed] [Google Scholar]
  • 16. Freedman DS, Khan LK, Dietz WH, et al. Relationship of childhood obesity to coronary heart disease risk factors in adulthood: the bogalusa heart study. Pediatrics. 2001;108(3):712‐718. [DOI] [PubMed] [Google Scholar]
  • 17. Loos RJF, Janssens A. Predicting polygenic obesity using genetic information. Cell Metab. 2017;25(3):535‐543. [DOI] [PubMed] [Google Scholar]
  • 18. Stunkard AJ, Foch TT, Hrubec Z. A twin study of human obesity. JAMA. 1986;256(1):51‐54. [PubMed] [Google Scholar]
  • 19. Maes HH, Neale MC, Eaves LJ. Genetic and environmental factors in relative body weight and human adiposity. Behav Genet. 1997;27(4):325‐351. [DOI] [PubMed] [Google Scholar]
  • 20. Loos RJ. The genetics of adiposity. Curr Opin Genet Dev. 2018;50:86‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Locke AE, Kahali B, Berndt SI, et 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]
  • 22. Turcot V, Lu Y, Highland HM, et al. Protein-altering variants associated with body mass index implicate pathways that control energy intake and expenditure in obesity. Nat Genet. 2018;50(1):26‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Khera AV, Chaffin M, Wade KH, et al. Polygenic prediction of weight and obesity trajectories from birth to adulthood. Cell. 2019;177(3):587‐596 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Akbari P, Gilani A, Sosina O, et al. Sequencing of 640,000 exomes identifies GPR75 variants associated with protection from obesity. Science. 2021;373(6550):eabf8683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Fahed AC, Wang M, Homburger JR, et al. Polygenic background modifies penetrance of monogenic variants for tier 1 genomic conditions. Nat Commun. 2020;11(1):3635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Damask A, Steg PG, Schwartz GG, et al. Patients with high genome-wide polygenic risk scores for coronary artery disease may receive greater clinical benefit from alirocumab treatment in the ODYSSEY OUTCOMES trial. Circulation. 2020;141(8):624‐636. [DOI] [PubMed] [Google Scholar]
  • 27. Westerterp KR, Speakman JR. Physical activity energy expenditure has not declined since the 1980s and matches energy expenditures of wild mammals. Int J Obes (Lond). 2008;32(8):1256‐1263. [DOI] [PubMed] [Google Scholar]
  • 28. Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine exposure in utero and early infancy. N Engl J Med. 1976;295(7):349‐353. [DOI] [PubMed] [Google Scholar]
  • 29. Roseboom TJ, Painter RC, van Abeelen AFM, et al. Hungry in the womb: what are the consequences? Lessons from the Dutch famine. Maturitas. 2011;70(2):141‐145. [DOI] [PubMed] [Google Scholar]
  • 30. Barker DJ. The fetal and infant origins of adult disease. BMJ. 1990;301(6761):1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Jones RH, Ozanne SE. Fetal programming of glucose-insulin metabolism. Mol Cell Endocrinol. 2009;297(1-2):4‐9. [DOI] [PubMed] [Google Scholar]
  • 32. Hales CN, Barker DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia. 1992;35(7):595‐601. [DOI] [PubMed] [Google Scholar]
  • 33. Lawlor DA, Smith GD, O'Callaghan M, et al. Epidemiologic evidence for the fetal overnutrition hypothesis: findings from the mater-university study of pregnancy and its outcomes. Am J Epidemiol. 2007;165(4):418‐424. [DOI] [PubMed] [Google Scholar]
  • 34. Alfaradhi MZ, Ozanne SE. Developmental programming in response to maternal overnutrition. Front Genet. 2011;2:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. George LA, Uthlaut AB, Long NM, et al. Different levels of overnutrition and weight gain during pregnancy have differential effects on fetal growth and organ development. Reprod Biol Endocrinol. 2010;8(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Gillman MW. Developmental origins of health and disease. N Engl J Med. 2005;353(17):1848‐1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Driscoll AK, Gregory ECW. Increases in prepregnancy obesity: United States, 2016–2019. NCHS Data Brief. 2020;(392):1‐8. [PubMed] [Google Scholar]
  • 38. Poston L, Caleyachetty R, Cnattingius S, et al. Preconceptional and maternal obesity: epidemiology and health consequences. Lancet Diabetes Endocrinol. 2016;4(12):1025‐1036. [DOI] [PubMed] [Google Scholar]
  • 39. Saucedo M, Esteves-Pereira AP, Pencolé L, et al. Understanding maternal mortality in women with obesity and the role of care they receive: a national case-control study. Int J Obes (Lond). 2021;45(1):258‐265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Lashen H, Fear K, Sturdee DW. Obesity is associated with increased risk of first trimester and recurrent miscarriage: matched case-control study. Hum Reprod. 2004;19(7):1644‐1646. [DOI] [PubMed] [Google Scholar]
  • 41. Weiss JL, Malone FD, Emig D, et al. Obesity, obstetric complications and cesarean delivery rate--a population-based screening study. Am J Obstet Gynecol. 2004;190(4):1091‐1097. [DOI] [PubMed] [Google Scholar]
  • 42. O'Brien TE, Ray JG, Chan WS. Maternal body mass index and the risk of preeclampsia: a systematic overview. Epidemiology. 2003;14(3):368‐374. [DOI] [PubMed] [Google Scholar]
  • 43. Vats H, Saxena R, Sachdeva MP, et al. Impact of maternal pre-pregnancy body mass index on maternal, fetal and neonatal adverse outcomes in the worldwide populations: A systematic review and meta-analysis. Obes Res Clin Pract. 2021;15(6):536‐545. [DOI] [PubMed] [Google Scholar]
  • 44. Najafi F, Hasani J, Izadi N, et al. The effect of prepregnancy body mass index on the risk of gestational diabetes mellitus: A systematic review and dose-response meta-analysis. Obes Rev. 2019;20(3):472‐486. [DOI] [PubMed] [Google Scholar]
  • 45. Ko CW, Beresford SAA, Schulte SJ, et al. Incidence, natural history, and risk factors for biliary sludge and stones during pregnancy. Hepatology. 2005;41(2):359‐365. [DOI] [PubMed] [Google Scholar]
  • 46. Marchi J, Berg M, Dencker A, et al. Risks associated with obesity in pregnancy, for the mother and baby: a systematic review of reviews. Obes Rev. 2015;16(8):621‐638. [DOI] [PubMed] [Google Scholar]
  • 47. Lahti-Pulkkinen M, Bhattacharya S, Wild SH, et al. Consequences of being overweight or obese during pregnancy on diabetes in the offspring: a record linkage study in Aberdeen, Scotland. Diabetologia. 2019;62(8):1412‐1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Reynolds RM, Allan KM, Raja EA, et al. Maternal obesity during pregnancy and premature mortality from cardiovascular event in adult offspring: follow-up of 1 323 275 person years. BMJ: British Medical Journal. 2013;347(aug13 1):f4539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Gaillard R, Steegers EAP, Duijts L, et al. Childhood cardiometabolic outcomes of maternal obesity during pregnancy. Hypertension. 2014;63(4):683‐691. [DOI] [PubMed] [Google Scholar]
  • 50. Nohr EA, Vaeth M, Baker JL, et al. Combined associations of prepregnancy body mass index and gestational weight gain with the outcome of pregnancy. Am J Clin Nutr. 2008;87(6):1750‐1759. [DOI] [PubMed] [Google Scholar]
  • 51. Yu Z, Han S, Zhu J, et al. Pre-pregnancy body mass index in relation to infant birth weight and offspring overweight/obesity: a systematic review and meta-analysis. PLoS One. 2013;8(4):e61627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Lausten-Thomsen U, Nielsen TRH, Thagaard IN, et al. Neonatal anthropometrics and body composition in obese children investigated by dual energy X-ray absorptiometry. Eur J Pediatr. 2014;173(5):623‐627. [DOI] [PubMed] [Google Scholar]
  • 53. Schellong K, Schulz S, Harder T, et al. Birth weight and long-term overweight risk: systematic review and a meta-analysis including 643,902 persons from 66 studies and 26 countries globally. PLoS One. 2012;7(10):e47776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Lake JK, Power C, Cole TJ. Child to adult body mass index in the 1958 British birth cohort: associations with parental obesity. Arch Dis Child. 1997;77(5):376‐381. [DOI] [PubMed] [Google Scholar]
  • 55. Catalano PM, Farrell K, Thomas A, et al. Perinatal risk factors for childhood obesity and metabolic dysregulation. Am J Clin Nutr. 2009;90(5):1303‐1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Linabery AM, Nahhas RW, Johnson W, et al. Stronger influence of maternal than paternal obesity on infant and early childhood body mass index: the fels longitudinal study. Pediatr Obes. 2013;8(3):159‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Danielzik S, Langnäse K, Mast M, et al. Impact of parental BMI on the manifestation of overweight 5–7 year old children. Eur J Nutr. 2002;41(3):132‐138. [DOI] [PubMed] [Google Scholar]
  • 58. Crozier SR, Inskip HM, Godfrey KM, et al. Weight gain in pregnancy and childhood body composition: findings from the southampton Women's Survey. Am J Clin Nutr. 2010;91(6):1745‐1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Hull HR, Thornton JC, Ji Y, et al. Higher infant body fat with excessive gestational weight gain in overweight women. Am J Obstet Gynecol. 2011;205(3):211.e1‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Derraik JGB, Maessen SE, Gibbins JD, et al. Large-for-gestational-age phenotypes and obesity risk in adulthood: a study of 195,936 women. Sci Rep. 2020;10(1):2157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Sorensen HT, Sabroe S, Rothman KJ, et al. Relation between weight and length at birth and body mass index in young adulthood: cohort study. BMJ. 1997;315(7116):1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Stettler N, Stallings VA, Troxel AB, et al. Weight gain in the first week of life and overweight in adulthood: a cohort study of European American subjects fed infant formula. Circulation. 2005;111(15):1897‐1903. [DOI] [PubMed] [Google Scholar]
  • 63. Andersen LG, Holst C, Michaelsen KF, et al. Weight and weight gain during early infancy predict childhood obesity: a case-cohort study. Int J Obes (Lond). 2012;36(10):1306‐1311. [DOI] [PubMed] [Google Scholar]
  • 64. Baird J, Fisher D, Lucas P, et al. Being big or growing fast: systematic review of size and growth in infancy and later obesity. BMJ. 2005;331(7522):929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Tzoulaki I, Sovio U, Pillas D, et al. Relation of immediate postnatal growth with obesity and related metabolic risk factors in adulthood: the northern Finland birth cohort 1966 study. Am J Epidemiol. 2010;171(9):989‐998. [DOI] [PubMed] [Google Scholar]
  • 66. Geserick M, Vogel M, Gausche R, et al. Acceleration of BMI in early childhood and risk of sustained obesity. N Engl J Med. 2018;379(14):1303‐1312. [DOI] [PubMed] [Google Scholar]
  • 67. Evensen E, Emaus N, Kokkvoll A, et al. The relation between birthweight, childhood body mass index, and overweight and obesity in late adolescence: a longitudinal cohort study from Norway, the tromsø study, fit futures. BMJ Open. 2017;7(6):e015576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Simmonds M, Llewellyn A, Owen CG, et al. Predicting adult obesity from childhood obesity: a systematic review and meta-analysis. Obes Rev. 2016;17(2):95‐107. [DOI] [PubMed] [Google Scholar]
  • 69. Geng T, Smith CE, Li C, et al. Childhood BMI and adult type 2 diabetes, coronary artery diseases, chronic kidney disease, and cardiometabolic traits: A Mendelian randomization analysis. Diabetes Care. 2018;41(5):1089‐1096. [DOI] [PubMed] [Google Scholar]
  • 70. Fang X, Zuo J, Zhou J, et al. Childhood obesity leads to adult type 2 diabetes and coronary artery diseases: A 2-sample Mendelian randomization study. Medicine (Baltimore). 2019;98(32):e16825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Catalano PM, Thomas A, Huston-Presley L, et al. Increased fetal adiposity: a very sensitive marker of abnormal in utero development. Am J Obstet Gynecol. 2003;189(6):1698‐1704. [DOI] [PubMed] [Google Scholar]
  • 72. Wiechers C, Balles LS, Kirchhof S, et al. Body composition in term offspring after maternal gestational diabetes does not predict postnatal hypoglycemia. BMC Pediatr. 2021;21(1):111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ehrlich SF, Crites YM, Hedderson MM, et al. The risk of large for gestational age across increasing categories of pregnancy glycemia. Am J Obstet Gynecol. 2011;204(3):240.e1‐240.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Herath MP, Beckett JM, Hills AP, et al. Gestational diabetes Mellitus and infant adiposity at birth: A systematic review and meta-analysis of therapeutic interventions. J Clin Med. 2021;10(4):835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Lindsay RS, Nelson SM, Walker JD, et al. Programming of adiposity in offspring of mothers with type 1 diabetes at age 7 years. Diabetes Care. 2010;33(5):1080‐1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Lawlor DA, Lichtenstein P, Långström N. Association of maternal diabetes mellitus in pregnancy with offspring adiposity into early adulthood: sibling study in a prospective cohort of 280,866 men from 248,293 families. Circulation. 2011;123(3):258‐265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Crowther CA, Hiller JE, Moss JR, et al. Effect of treatment of gestational diabetes Mellitus on pregnancy outcomes. N Engl J Med. 2005;352(24):2477‐2486. [DOI] [PubMed] [Google Scholar]
  • 78. Landon MB, Spong CY, Thom E, et al. A multicenter, randomized trial of treatment for mild gestational diabetes. N Engl J Med. 2009;361(14):1339‐1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Cyganek K, Skupien J, Katra B, et al. Risk of macrosomia remains glucose-dependent in a cohort of women with pregestational type 1 diabetes and good glycemic control. Endocrine. 2017;55(2):447‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Evers IM, de Valk H, Mol B, et al. Macrosomia despite good glycaemic control in type I diabetic pregnancy; results of a nationwide study in The Netherlands. Diabetologia. 2002;45(11):1484‐1489. [DOI] [PubMed] [Google Scholar]
  • 81. Hernandez TL. Glycemic targets in pregnancies affected by diabetes: historical perspective and future directions. Curr Diab Rep. 2015;15(1):565. [DOI] [PubMed] [Google Scholar]
  • 82. Langer O, Yogev Y, Xenakis EMJ, et al. Overweight and obese in gestational diabetes: the impact on pregnancy outcome. Am J Obstet Gynecol. 2005;192(6):1768‐1776. [DOI] [PubMed] [Google Scholar]
  • 83. Dabelea D, Hanson RL, Lindsay RS, et al. Intrauterine exposure to diabetes conveys risks for type 2 diabetes and obesity: a study of discordant sibships. Diabetes. 2000;49(12):2208‐2211. [DOI] [PubMed] [Google Scholar]
  • 84. Portha B, Grandjean V, Movassat J. Mother or father: who is in the front line? Mechanisms underlying the non-genomic transmission of obesity/diabetes via the maternal or the paternal line. Nutrients. 2019;11(2):233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Adam PAJ, Teramo K, Raiha N, et al. Human fetal insulin metabolism early in gestation: response to acute elevation of the fetal glucose concentration and placental transfer of human insulin-I-131. Diabetes. 1969;18(6):409‐416. [DOI] [PubMed] [Google Scholar]
  • 86. Bishop KC, Harris BS, Boyd BK, et al. Pharmacologic treatment of diabetes in pregnancy. Obstet Gynecol Surv. 2019;74(5):289‐297. [DOI] [PubMed] [Google Scholar]
  • 87. Pedersen, J. Diabetes and Pregnancy. Blood Sugar of Newborn Infants. A thesis. Danish Science Press:Copenhagen; 1952.
  • 88. HAPO Study Cooperative Research Group, Metzger BE, Lowe LP, et al. Hyperglycemia and adverse pregnancy outcomes. N Engl J Med. 2008;358(19):1991‐2002. [DOI] [PubMed] [Google Scholar]
  • 89. Casson IF, Clarke CA, Howard CV, et al. Outcomes of pregnancy in insulin dependent diabetic women: results of a five year population cohort study. BMJ. 1997;315(7103):275‐278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Dunne F, Brydon P, Smith K, et al. Pregnancy in women with type 2 diabetes: 12 years outcome data 1990–2002. Diabet Med. 2003;20(9):734‐738. [DOI] [PubMed] [Google Scholar]
  • 91. Arendt LH, Pedersen LH, Pedersen L, et al. Glycemic control in pregnancies complicated by Pre-existing diabetes Mellitus and congenital malformations: A danish population-based study. Clin Epidemiol. 2021;13:615‐626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zhang T-N, Huang X-M, Zhao X-Y, et al. Risks of specific congenital anomalies in offspring of women with diabetes: A systematic review and meta-analysis of population-based studies including over 80 million births. PLoS Med. 2022;19(2):e1003900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Freinkel N. Banting lecture 1980. Of pregnancy and progeny. Diabetes. 1980;29(12):1023‐1035. [DOI] [PubMed] [Google Scholar]
  • 94. Susa JB, Neave C, Sehgal P, et al. Chronic hyperinsulinemia in the fetal rhesus monkey: effects of physiologic hyperinsulinemia on fetal growth and composition. Diabetes. 1984;33(7):656‐660. [DOI] [PubMed] [Google Scholar]
  • 95. Catlin EA, Cha CJ, Oh W. Postnatal growth and fatty acid synthesis in overgrown rat pups induced by fetal hyperinsulinemia. Metab Clin Exp. 1985;34(12):1110‐1114. [DOI] [PubMed] [Google Scholar]
  • 96. Ogata ES, Collins JW Jr, Finley S. Insulin injection in the fetal rat: accelerated intrauterine growth and altered fetal and neonatal glucose homeostasis. Metab Clin Exp. 1988;37(7):649‐655. [DOI] [PubMed] [Google Scholar]
  • 97. Ogata ES, Paul RI, Finley SL. Limited maternal fuel availability due to hyperinsulinemia retards fetal growth and development in the rat. Pediatr Res. 1987;22(4):432‐437. [DOI] [PubMed] [Google Scholar]
  • 98. Lee I-L, Barr ELM, Longmore D, et al. Cord blood metabolic markers are strong mediators of the effect of maternal adiposity on fetal growth in pregnancies across the glucose tolerance spectrum: the PANDORA study. Diabetologia. 2020;63(3):497‐507. [DOI] [PubMed] [Google Scholar]
  • 99. Josefson JL, Scholtens DM, Kuang A, et al. Newborn adiposity and cord blood C-peptide as mediators of the maternal metabolic environment and childhood adiposity. Diabetes Care. 2021;44(5):1194‐1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Byrne MM, Sturis J, Clément K, et al. Insulin secretory abnormalities in subjects with hyperglycemia due to glucokinase mutations. J Clin Invest. 1994;93(3):1120‐1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Hattersley AT, Beards F, Ballantyne E, et al. Mutations in the glucokinase gene of the fetus result in reduced birth weight. Nat Genet. 1998;19(3):268‐270. [DOI] [PubMed] [Google Scholar]
  • 102. O'Tierney-Ginn P, Presley L, Myers S, et al. Placental growth response to maternal insulin in early pregnancy. J Clin Endocrinol Metab. 2015;100(1):159‐165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Tanaka K, Yamada K, Matsushima M, et al. Increased maternal insulin resistance promotes placental growth and decreases placental efficiency in pregnancies with obesity and gestational diabetes mellitus. J Obstet Gynaecol Res. 2018;44(1):74‐80. [DOI] [PubMed] [Google Scholar]
  • 104. Shields BM, Spyer G, Slingerland AS, et al. Mutations in the glucokinase gene of the fetus result in reduced placental weight. Diabetes Care. 2008;31(4):753‐757. [DOI] [PubMed] [Google Scholar]
  • 105. Hiden U, Maier A, Bilban M, et al. Insulin control of placental gene expression shifts from mother to foetus over the course of pregnancy. Diabetologia. 2006;49(1):123‐131. [DOI] [PubMed] [Google Scholar]
  • 106. Lassance L, Miedl H, Absenger M, et al. Hyperinsulinemia stimulates angiogenesis of human fetoplacental endothelial cells: a possible role of insulin in placental hypervascularization in diabetes mellitus. J Clin Endocrinol Metab. 2013;98(9):E1438‐E1447. [DOI] [PubMed] [Google Scholar]
  • 107. Boerschmann H, Pflüger M, Henneberger L, et al. Prevalence and predictors of overweight and insulin resistance in offspring of mothers with gestational diabetes mellitus. Diabetes Care. 2010;33(8):1845‐1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Villamor E, Cnattingius S. Interpregnancy weight change and risk of adverse pregnancy outcomes: a population-based study. Lancet. 2006;368(9542):1164‐1170. [DOI] [PubMed] [Google Scholar]
  • 109. Teulings NEWD, Masconi KL, Ozanne SE, et al. Effect of interpregnancy weight change on perinatal outcomes: systematic review and meta-analysis. BMC Pregnancy Childbirth. 2019;19(1):386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Timmermans YEG, van de Kant KDG, Oosterman EO, et al. The impact of interpregnancy weight change on perinatal outcomes in women and their children: A systematic review and meta-analysis. Obes Rev. 2020;21(3):e12974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Ziauddeen N, Wilding S, Roderick PJ, et al. Is maternal weight gain between pregnancies associated with risk of large-for-gestational age birth? Analysis of a UK population-based cohort. BMJ Open. 2019;9(7):e026220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Kral JG, Biron S, Simard S, et al. Large maternal weight loss from obesity surgery prevents transmission of obesity to children who were followed for 2 to 18 years. Pediatrics. 2006;118(6):e1644‐e1649. [DOI] [PubMed] [Google Scholar]
  • 113. Smith J, Cianflone K, Biron S, et al. Effects of maternal surgical weight loss in mothers on intergenerational transmission of obesity. J Clin Endocrinol Metab. 2009;94(11):4275‐4283. [DOI] [PubMed] [Google Scholar]
  • 114. Barisione M, Carlini F, Gradaschi R, et al. Body weight at developmental age in siblings born to mothers before and after surgically induced weight loss. Surg Obes Relat Dis. 2012;8(4):387‐391. [DOI] [PubMed] [Google Scholar]
  • 115. Ibiebele I, Gallimore F, Schnitzler M, et al. Perinatal outcomes following bariatric surgery between a first and second pregnancy: a population data linkage study. BJOG. 2020;127(3):345‐354. [DOI] [PubMed] [Google Scholar]
  • 116. Guénard F, Deshaies Y, Cianflone K, et al. Differential methylation in glucoregulatory genes of offspring born before vs. After maternal gastrointestinal bypass surgery. Proc Natl Acad Sci U S A. 2013;110(28):11439‐11444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Howie GJ, Sloboda DM, Vickers MH. Maternal undernutrition during critical windows of development results in differential and sex-specific effects on postnatal adiposity and related metabolic profiles in adult rat offspring. Br J Nutr. 2012;108(2):298‐307. [DOI] [PubMed] [Google Scholar]
  • 118. Desai M, Gayle D, Babu J, et al. Programmed obesity in intrauterine growth-restricted newborns: modulation by newborn nutrition. Am J Physiol Regul Integr Comp Physiol. 2005;288(1):R91‐R96. [DOI] [PubMed] [Google Scholar]
  • 119. Vickers MH, Breier BH, Cutfield WS, et al. Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am J Physiol Endocrinol Metab. 2000;279(1):E83‐E87. [DOI] [PubMed] [Google Scholar]
  • 120. Cripps R, Martin-Gronert M, Archer Z, et al. Programming of hypothalamic neuropeptide gene expression in rats by maternal dietary protein content during pregnancy and lactation. Clin Sci (Lond). 2009;117(2):85‐93. [DOI] [PubMed] [Google Scholar]
  • 121. Delahaye F, Breton C, Risold P-Y, et al. Maternal perinatal undernutrition drastically reduces postnatal leptin surge and affects the development of arcuate nucleus proopiomelanocortin neurons in neonatal male rat pups. Endocrinology. 2008;149(2):470‐475. [DOI] [PubMed] [Google Scholar]
  • 122. Yura S, Itoh H, Sagawa N, et al. Role of premature leptin surge in obesity resulting from intrauterine undernutrition. Cell Metab. 2005;1(6):371‐378. [DOI] [PubMed] [Google Scholar]
  • 123. Aubert R, Suquet J-P, Lemonnier D. Long-Term morphological and metabolic effects of early under- and over-nutrition in mice. J Nutr. 1980;110(4):649‐661. [DOI] [PubMed] [Google Scholar]
  • 124. Marangon PB, Mecawi AS, Antunes-Rodrigues J, et al. Perinatal over- and underfeeding affect hypothalamic leptin and ghrelin neuroendocrine responses in adult rats. Physiol Behav. 2020;215:112793. [DOI] [PubMed] [Google Scholar]
  • 125. Woodall SM, Breier BH, Johnston BM, et al. A model of intrauterine growth retardation caused by chronic maternal undernutrition in the rat: effects on the somatotrophic axis and postnatal growth. J Endocrinol. 1996;150(2):231‐242. [DOI] [PubMed] [Google Scholar]
  • 126. Garofano A, Czernichow P, Bréant B. In utero undernutrition impairs rat beta-cell development. Diabetologia. 1997;40(10):1231‐1234. [DOI] [PubMed] [Google Scholar]
  • 127. Jimenez-Chillaron JC, Hernandez-Valencia M, Reamer C, et al. Beta-cell secretory dysfunction in the pathogenesis of low birth weight-associated diabetes: a murine model. Diabetes. 2005;54(3):702‐711. [DOI] [PubMed] [Google Scholar]
  • 128. Ozanne SE, Olsen GS, Hansen LL, et al. Early growth restriction leads to down regulation of protein kinase C zeta and insulin resistance in skeletal muscle. Journal of Endocrinology. 2003;177(2):235‐241. [DOI] [PubMed] [Google Scholar]
  • 129. Fernandez-Twinn DS, Wayman A, Ekizoglou S, et al. Maternal protein restriction leads to hyperinsulinemia and reduced insulin-signaling protein expression in 21-mo-old female rat offspring. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2005;288(2):R368‐R373. [DOI] [PubMed] [Google Scholar]
  • 130. Berends LM, Fernandez-Twinn DS, Martin-Gronert MS, et al. Catch-up growth following intra-uterine growth-restriction programmes an insulin-resistant phenotype in adipose tissue. Int J Obes (Lond). 2013;37(8):1051‐1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Desai M, Gayle D, Babu J, et al. The timing of nutrient restriction during rat pregnancy/lactation alters metabolic syndrome phenotype. Am J Obstet Gynecol. 2007;196(6):555.e1‐555.e5557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Jimenez-Chillaron JC, Hernandez-Valencia M, Lightner A, et al. Reductions in caloric intake and early postnatal growth prevent glucose intolerance and obesity associated with low birthweight. Diabetologia. 2006;49(8):1974‐1984. [DOI] [PubMed] [Google Scholar]
  • 133. Bieswal F, Ahn M-T, Reusens B, et al. The importance of catch-up growth after early malnutrition for the programming of obesity in male rat. Obesity. 2006;14(8):1330‐1343. [DOI] [PubMed] [Google Scholar]
  • 134. Garg M, Thamotharan M, Dai Y, et al. Early postnatal caloric restriction protects adult male intrauterine growth–restricted offspring from obesity. Diabetes. 2012;61(6):1391‐1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Desai M, Babu J, Ross MG. Programmed metabolic syndrome: prenatal undernutrition and postweaning overnutrition. Am J Physiol Regul Integr Comp Physiol. 2007;293(6):R2306‐R2314. [DOI] [PubMed] [Google Scholar]
  • 136. Ozanne SE, Lewis R, Jennings BJ, et al. Early programming of weight gain in mice prevents the induction of obesity by a highly palatable diet. Clin Sci (Lond). 2004;106(2):141‐145. [DOI] [PubMed] [Google Scholar]
  • 137. Sutton GM, Centanni AV, Butler AA. Protein malnutrition during pregnancy in C57BL/6J mice results in offspring with altered circadian physiology before obesity. Endocrinology. 2010;151(4):1570‐1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Bol VV, Delattre A-I, Reusens B, et al. Forced catch-up growth after fetal protein restriction alters the adipose tissue gene expression program leading to obesity in adult mice. Am J Physiol Regul Integr Comp Physiol. 2009;297(2):R291‐R299. [DOI] [PubMed] [Google Scholar]
  • 139. Coupé B, Amarger V, Grit I, et al. Nutritional programming affects hypothalamic organization and early response to leptin. Endocrinology. 2010;151(2):702‐713. [DOI] [PubMed] [Google Scholar]
  • 140. Zambrano E, Bautista CJ, Deás M, et al. A low maternal protein diet during pregnancy and lactation has sex- and window of exposure-specific effects on offspring growth and food intake, glucose metabolism and serum leptin in the rat. J Physiol (Lond). 2006;571(1):221‐230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Stocker CJ, Wargent ET, Martin-Gronert MS, et al. Leanness in postnatally nutritionally programmed rats is associated with increased sensitivity to leptin and a melanocortin receptor agonist and decreased sensitivity to neuropeptide Y. Int J Obes (Lond). 2012;36(8):1040‐1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Ikenasio-Thorpe BA, Breier BH, Vickers MH, et al. Prenatal influences on susceptibility to diet-induced obesity are mediated by altered neuroendocrine gene expression. J Endocrinol. 2007;193(1):31‐37. [DOI] [PubMed] [Google Scholar]
  • 143. Alexandre-Gouabau M-C, Courant F, Le Gall G, et al. Offspring metabolomic response to maternal protein restriction in a rat model of intrauterine growth restriction (IUGR). J Proteome Res. 2011;10(7):3292‐3302. [DOI] [PubMed] [Google Scholar]
  • 144. Krechowec SO, Vickers M, Gertler A, et al. Prenatal influences on leptin sensitivity and susceptibility to diet-induced obesity. J Endocrinol. 2006;189(2):355‐363. [DOI] [PubMed] [Google Scholar]
  • 145. Desai M, Gayle D, Han G, et al. Programmed hyperphagia due to reduced anorexigenic mechanisms in intrauterine growth-restricted offspring. Reprod Sci. 2007;14(4):329‐337. [DOI] [PubMed] [Google Scholar]
  • 146. Bautista CJ, Boeck L, Larrea F, et al. Effects of a maternal low protein isocaloric diet on milk leptin and progeny Serum leptin concentration and appetitive behavior in the first 21 days of neonatal life in the rat. Pediatr Res. 2008;63(4):358‐363. [DOI] [PubMed] [Google Scholar]
  • 147. López M, Seoane LM, Tovar S, et al. A possible role of neuropeptide Y, agouti-related protein and leptin receptor isoforms in hypothalamic programming by perinatal feeding in the rat. Diabetologia. 2005;48(1):140‐148. [DOI] [PubMed] [Google Scholar]
  • 148. Kappeler L, De Magalhaes Filho C, Leneuve P, et al. Early postnatal nutrition determines somatotropic function in mice. Endocrinology. 2009;150(1):314‐323. [DOI] [PubMed] [Google Scholar]
  • 149. Skowronski AA, Shaulson ED, Leibel RL, et al. The postnatal leptin surge in mice is variable in both time and intensity and reflects nutritional status. Int J Obes. 2021;46:39‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Zhang L, Morgan DG, Clapham JC, et al. Factors predicting nongenetic variability in body weight gain induced by a high-fat diet in inbred C57BL/6J mice. Obesity (Silver Spring). 2012;20(6):1179‐1188. [DOI] [PubMed] [Google Scholar]
  • 151. Remmers F, Fodor M, Delemarre-van de Waal HA. Neonatal food restriction permanently alters rat body dimensions and energy intake. Physiol Behav. 2008;95(1-2):208‐215. [DOI] [PubMed] [Google Scholar]
  • 152. Samuelsson A-M, Matthews PA, Argenton M, et al. Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension. 2008;51(2):383‐392. [DOI] [PubMed] [Google Scholar]
  • 153. Gomes RM, Bueno FG, Schamber CR, et al. Maternal diet-induced obesity during suckling period programs offspring obese phenotype and hypothalamic leptin/insulin resistance. J Nutr Biochem. 2018;61:24‐32. [DOI] [PubMed] [Google Scholar]
  • 154. Ashino NG, Saito KN, Souza FD, et al. Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J Nutr Biochem. 2012;23(4):341‐348. [DOI] [PubMed] [Google Scholar]
  • 155. Elahi MM, Cagampang FR, Mukhtar D, et al. Long-term maternal high-fat feeding from weaning through pregnancy and lactation predisposes offspring to hypertension, raised plasma lipids and fatty liver in mice. Br J Nutr. 2009;102(4):514‐519. [DOI] [PubMed] [Google Scholar]
  • 156. Masuyama H, Hiramatsu Y. Additive effects of maternal high fat diet during lactation on mouse offspring. PLoS One. 2014;9(3):e92805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Franco JG, Fernandes TP, Rocha CPD, et al. Maternal high-fat diet induces obesity and adrenal and thyroid dysfunction in male rat offspring at weaning. J Physiol. 2012;590(21):5503‐5518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Bayol SA, Farrington SJ, Stickland NC. A maternal ‘junk food’ diet in pregnancy and lactation promotes an exacerbated taste for ‘junk food’ and a greater propensity for obesity in rat offspring. Br J Nutr. 2007;98(04):843‐851. [DOI] [PubMed] [Google Scholar]
  • 159. Sun B, Purcell RH, Terrillion CE, et al. Maternal high-fat diet during gestation or suckling differentially affects offspring leptin sensitivity and obesity. Diabetes. 2012;61(11):2833‐2841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Vogt M, Paeger L, Hess S, et al. Neonatal insulin action impairs hypothalamic neurocircuit formation in response to maternal high-fat feeding. Cell. 2014;156(3):495‐509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Huang Y, Osorio Mendoza J, Li M, et al. Impact of graded maternal dietary fat content on offspring susceptibility to high-fat diet in mice. Obesity (Silver Spring). 2021;29(12):2055‐2067. [DOI] [PubMed] [Google Scholar]
  • 162. Huang Y, Mendoza JO, Hambly C, et al. Limits to sustained energy intake. XXXI. Effect of graded levels of dietary fat on lactation performance in Swiss mice. J Exp Biol. 2020;223(Pt 10):jeb221911. [DOI] [PubMed] [Google Scholar]
  • 163. Chahoud I, Paumgartten FJR. Influence of litter size on the postnatal growth of rat pups: is there a rationale for litter-size standardization in toxicity studies? Environ Res. 2009;109(8):1021‐1027. [DOI] [PubMed] [Google Scholar]
  • 164. Mistry AM, Swick A, Romsos DR. Leptin alters metabolic rates before acquisition of its anorectic effect in developing neonatal mice. Am J Physiol. 1999;277(3):R742‐R747. [DOI] [PubMed] [Google Scholar]
  • 165. Proulx K, Richard D, Walker CD. Leptin regulates appetite-related neuropeptides in the hypothalamus of developing rats without affecting food intake. Endocrinology. 2002;143(12):4683‐4692. [DOI] [PubMed] [Google Scholar]
  • 166. Skowronski AA, LeDuc CA, Foo KS, et al. Physiological consequences of transient hyperleptinemia during discrete developmental periods on body weight in mice. Sci Transl Med. 2020;12(524):eaax6629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Glavas MM, Kirigiti MA, Xiao XQ, et al. Early overnutrition results in early-onset arcuate leptin resistance and increased sensitivity to high-fat diet. Endocrinology. 2010;151(4):1598‐1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Liu Z, Lim CY, Su MY-F, et al. Neonatal overnutrition in mice exacerbates high-fat diet-induced metabolic perturbations. J Endocrinol. 2013;219(2):131‐143. [DOI] [PubMed] [Google Scholar]
  • 169. Purcell RH, Sun B, Pass LL, et al. Maternal stress and high-fat diet effect on maternal behavior, milk composition, and pup ingestive behavior. Physiol Behav. 2011;104(3):474‐479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Bautista CJ, Montaño S, Ramirez V, et al. Changes in milk composition in obese rats consuming a high-fat diet. Br J Nutr. 2016;115(3):538‐546. [DOI] [PubMed] [Google Scholar]
  • 171. Del Prado M, Delgado G, Villalpando S. Maternal lipid intake during pregnancy and lactation alters milk composition and production and litter growth in rats. J Nutr. 1997;127(3):458‐462. [DOI] [PubMed] [Google Scholar]
  • 172. Rolls BA, Gurr MI, Van Duijvenvoorde PM, et al. Lactation in lean and obese rats: effect of cafeteria feeding and of dietary obesity on milk composition. Physiol Behav. 1986;38(2):185‐190. [DOI] [PubMed] [Google Scholar]
  • 173. de Souza Rodrigues Cunha AC, Pereira RO, dos Santos Pereira MJ, et al. Long-term effects of overfeeding during lactation on insulin secretion--the role of GLUT-2. J Nutr Biochem. 2009;20(6):435‐442. [DOI] [PubMed] [Google Scholar]
  • 174. Xavier JLP, Scomparin DX, Pontes CC, et al. Litter size reduction induces metabolic and histological adjustments in dams throughout lactation with early effects on offspring. An Acad Bras Cienc. 2019;91(1):e20170971. [DOI] [PubMed] [Google Scholar]
  • 175. Sefcikova Z, Bujňáková D, Raček L’, et al. Developmental changes in gut microbiota and enzyme activity predict obesity risk in rats arising from reduced nests. Physiol Res. 2011;60(2):337‐346. [DOI] [PubMed] [Google Scholar]
  • 176. Fiorotto ML, Burrin DG, Perez M, et al. Intake and use of milk nutrients by rat pups suckled in small, medium, or large litters. Am J Physiol. 1991;260(6 Pt 2):R1104‐R1113. [DOI] [PubMed] [Google Scholar]
  • 177. Ahima RS, Prabakaran D, Flier JS. Postnatal leptin surge and regulation of circadian rhythm of leptin by feeding. Implications for energy homeostasis and neuroendocrine function. J Clin Invest. 1998;101(5):1020‐1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Kirk SL, Samuelsson A-M, Argenton M, et al. Maternal obesity induced by diet in rats permanently influences central processes regulating food intake in offspring. PLoS One. 2009;4(6):e5870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Dearden L, Balthasar N. Sexual dimorphism in offspring glucose-sensitive hypothalamic gene expression and physiological responses to maternal high-fat diet feeding. Endocrinology. 2014;155(6):2144‐2154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Belgardt BF, Bruning JC. CNS Leptin and insulin action in the control of energy homeostasis. Ann N Y Acad Sci. 2010;1212(1):97‐113. [DOI] [PubMed] [Google Scholar]
  • 181. Bouret SG. Role of early hormonal and nutritional experiences in shaping feeding behavior and hypothalamic development. J Nutr. 2010;140(3):653‐657. [DOI] [PubMed] [Google Scholar]
  • 182. Cowley MA, Smart JL, Rubinstein M, et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature. 2001;411(6836):480‐484. [DOI] [PubMed] [Google Scholar]
  • 183. Luquet S, Perez FA, Hnasko TS, et al. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science. 2005;310(5748):683‐685. [DOI] [PubMed] [Google Scholar]
  • 184. Clancy B, Darlington RB, Finlay BL. Translating developmental time across mammalian species. Neuroscience. 2001;105(1):7‐17. [DOI] [PubMed] [Google Scholar]
  • 185. Bayer SA, Altman J, Russo RJ, et al. Timetables of neurogenesis in the human brain based on experimentally determined patterns in the rat. Neurotoxicology. 1993;14(1):83‐144. [PubMed] [Google Scholar]
  • 186. Cardoso-Moreira M, Halbert J, Valloton D, et al. Gene expression across mammalian organ development. Nature. 2019;571(7766):505‐509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Altman J, Bayer SA. The development of the rat hypothalamus. Adv Anat Embryol Cell Biol. 1986;100:1‐178. [PubMed] [Google Scholar]
  • 188. Shimada M, Nakamura T. Time of neuron origin in mouse hypothalamic nuclei. Exp Neurol. 1973;41(1):163‐173. [DOI] [PubMed] [Google Scholar]
  • 189. Ishii Y, Bouret SG. Embryonic birthdate of hypothalamic leptin-activated neurons in mice. Endocrinology. 2012;153(8):3657‐3667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Udagawa J, Hatta T, Naora H, et al. Expression of the long form of leptin receptor (ob-rb) mRNA in the brain of mouse embryos and newborn mice. Brain Res. 2000;868(2):251‐258. [DOI] [PubMed] [Google Scholar]
  • 191. Klein MO, MacKay H, Edwards A, et al. POMC And NPY mRNA expression during development is increased in rat offspring brain from mothers fed with a high fat diet. Int J Dev Neurosci. 2018;64(1):14‐20. [DOI] [PubMed] [Google Scholar]
  • 192. Udagawa J, Hashimoto R, Suzuki H, et al. The role of leptin in the development of the cerebral cortex in mouse embryos. Endocrinology. 2006;147(2):647‐658. [DOI] [PubMed] [Google Scholar]
  • 193. Steppan CM, Swick AG. A role for leptin in brain development. Biochem Biophys Res Commun. 1999;256(3):600‐602. [DOI] [PubMed] [Google Scholar]
  • 194. Bereiter DA, Jeanrenaud B. Altered neuroanatomical organization in the central nervous system of the genetically obese (ob/ob) mouse. Brain Res. 1979;165(2):249‐260. [DOI] [PubMed] [Google Scholar]
  • 195. Desai M, Li T, Ross MG. Fetal hypothalamic neuroprogenitor cell culture: preferential differentiation paths induced by leptin and insulin. Endocrinology. 2011;152(8):3192‐3201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Padilla SL, Carmody JS, Zeltser LM. Pomc-expressing progenitors give rise to antagonistic neuronal populations in hypothalamic feeding circuits. Nat Med. 2010;16(4):403‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Carmody JS, Wan P, Accili D, Zeltser LM, Leibel RL. Respective contributions of maternal insulin resistance and diet to metabolic and hypothalamic phenotypes of progeny. Obesity (Silver Spring). 2011;19(3):492‐499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004;304(5667):108‐110. [DOI] [PubMed] [Google Scholar]
  • 199. Bouret SG, Draper SJ, Simerly RB. Formation of projection pathways from the arcuate nucleus of the hypothalamus to hypothalamic regions implicated in the neural control of feeding behavior in mice. J Neurosci. 2004;24(11):2797‐2805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Bouyer K, Simerly RB. Neonatal leptin exposure specifies innervation of presympathetic hypothalamic neurons and improves the metabolic status of leptin-deficient mice. J Neurosci. 2013;33(2):840‐851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Baquero AF, de Solis AJ, Lindsley SR, et al. Developmental switch of leptin signaling in arcuate nucleus neurons. J Neurosci. 2014;34(30):9982‐9994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Hayes M, Skibicka K, Leichner T, et al. Endogenous leptin signaling in the caudal nucleus tractus Solitarius and area postrema is required for energy balance regulation. Cell Metab. 2016;23(4):744. [DOI] [PubMed] [Google Scholar]
  • 203. Hayes MR, Skibicka KP, Leichner TM, et al. Endogenous leptin signaling in the caudal nucleus tractus solitarius and area postrema is required for energy balance regulation. Cell Metab. 2010;11(1):77‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Huo L, Gamber KM, Grill HJ, Bjørbæk C. Divergent leptin signaling in proglucagon neurons of the nucleus of the solitary tract in mice and rats. Endocrinology. 2008;149(2):492‐497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Grill HJ, Schwartz MW, Kaplan JM, Foxhall JS, Breininger J, Baskin DG. Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology. 2002;143(1):239‐246. [DOI] [PubMed] [Google Scholar]
  • 206. Larsen PJ, Tang-Christensen M, Holst JJ, Ørskov C. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience. 1997;77(1):257‐270. [DOI] [PubMed] [Google Scholar]
  • 207. Garfield AS, Patterson C, Skora S, et al. Neurochemical characterization of body weight-regulating leptin receptor neurons in the nucleus of the solitary tract. Endocrinology. 2012;153(10):4600‐4607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Cheng W, Ndoka E, Hutch C, et al. Leptin receptor–expressing nucleus tractus solitarius neurons suppress food intake independently of GLP1 in mice. JCI Insight. 2020;5(7):e134359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Biddinger JE, Lazarenko RM, Scott MM, Simerly R, . Leptin suppresses development of GLP-1 inputs to the paraventricular nucleus of the hypothalamus. Elife. 2020;9:e59857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Berglund ED, Vianna CR, Donato J, et al. Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J Clin Invest. 2012;122(3):1000‐1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Palmiter RD. Is dopamine a physiologically relevant mediator of feeding behavior? Trends Neurosci. 2007;30(8):375‐381. [DOI] [PubMed] [Google Scholar]
  • 212. Geiger BM, Haburcak M, Avena NM, Moyer MC, Hoebel BG, Pothos EN. Deficits of mesolimbic dopamine neurotransmission in rat dietary obesity. Neuroscience. 2009;159(4):1193‐1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Demos KE, Heatherton TF, Kelley WM. Individual differences in nucleus accumbens activity to food and sexual images predict weight gain and sexual behavior. J Neurosci. 2012;32(16):5549‐5552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Shapiro ALB, Johnson SL, Sutton B, Legget KT, Dabelea D, Tregellas JR. Eating in the absence of hunger in young children is related to brain reward network hyperactivity and reduced functional connectivity in executive control networks. Pediatr Obes. 2019;14(6):e12502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Figlewicz DP, Evans SB, Murphy J, Hoen M, Baskin DG. Expression of receptors for insulin and leptin in the ventral tegmental area/substantia nigra (VTA/SN) of the rat. Brain Res. 2003;964(1):107‐115. [DOI] [PubMed] [Google Scholar]
  • 216. Hommel JD, Trinko R, Sears RM, et al. Leptin receptor signaling in midbrain dopamine neurons regulates feeding. Neuron. 2006;51(6):801‐810. [DOI] [PubMed] [Google Scholar]
  • 217. Leshan RL, Opland DM, Louis GW, et al. Ventral tegmental area leptin receptor neurons specifically project to and regulate cocaine- and amphetamine-regulated transcript neurons of the extended central amygdala. J Neurosci. 2010;30(16):5713‐5723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Fulton S, Pissios P, Manchon R, et al. Leptin regulation of the mesoaccumbens dopamine pathway. Neuron. 2006;51(6):811‐822. [DOI] [PubMed] [Google Scholar]
  • 219. Davis JF, Choi DL, Schurdak JD, et al. Leptin regulates energy balance and motivation through action at distinct neural circuits. Biol Psychiatry. 2011;69(7):668‐674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Morton GJ, Blevins JE, Kim F, Matsen M, Figlewicz DP. The action of leptin in the ventral tegmental area to decrease food intake is dependent on Jak-2 signaling. Am J Physiol Endocrinol Metab. 2009;297(1):E202‐E210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Leinninger GM, Jo Y-H, Leshan RL, et al. Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding. Cell Metab. 2009;10(2):89‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Omrani A, de Vrind VAJ, Lodder B, et al. Identification of novel neurocircuitry through which leptin targets multiple inputs to the dopamine system to reduce food reward seeking. Biol Psychiatry. 2021;90(12):843‐852. [DOI] [PubMed] [Google Scholar]
  • 223. Gjerde E, Long H, Richard D, Walker C-D. Developmental responses of the lateral hypothalamus to leptin in neonatal rats, and its implications for the development of functional connections with the ventral tegmental area. J Neuroendocrinol. 2016;28(3):12354. [DOI] [PubMed] [Google Scholar]
  • 224. Naef L, Gjerde E, Long H, Richard D, Walker C-D. Neonatal onset of leptin signalling in dopamine neurones of the ventral tegmental area in the rat. J Neuroendocrinol. 2014;26(12):835‐843. [DOI] [PubMed] [Google Scholar]
  • 225. Ong ZY, Muhlhausler BS. Maternal “junk-food” feeding of rat dams alters food choices and development of the mesolimbic reward pathway in the offspring. FASEB J. 2011;25(7):2167‐2179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Lippert RN, Hess S, Klemm P, et al. Maternal high-fat diet during lactation reprograms the dopaminergic circuitry in mice. J Clin Invest. 2020;130(7):3761‐3776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. de Oliveira Cravo C, Teixeira CV, Passos MCF, Dutra SCP, de Moura EG, Ramos C. Leptin treatment during the neonatal period is associated with higher food intake and adult body weight in rats. Horm Metab Res. 2002;34(7):400‐405. [DOI] [PubMed] [Google Scholar]
  • 228. Vickers MH, Gluckman PD, Coveny AH, et al. The effect of neonatal leptin treatment on postnatal weight gain in male rats is dependent on maternal nutritional status during pregnancy. Endocrinology. 2008;149(4):1906‐1913. [DOI] [PubMed] [Google Scholar]
  • 229. Kim JG, Suyama S, Koch M, et al. Leptin signaling in astrocytes regulates hypothalamic neuronal circuits and feeding. Nat Neurosci. 2014;17(7):908‐910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Bandeira F, Lent R, Herculano-Houzel S. Changing numbers of neuronal and non-neuronal cells underlie postnatal brain growth in the rat. Proc Natl Acad Sci U S A. 2009;106(33):14108‐14113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Clarke LE, Barres BA. Emerging roles of astrocytes in neural circuit development. Nat Rev Neurosci. 2013;14(5):311‐321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Nedergaard M, Ransom B, Goldman SA. New roles for astrocytes: redefining the functional architecture of the brain. Trends Neurosci. 2003;26(10):523‐530. [DOI] [PubMed] [Google Scholar]
  • 233. Leloup C, Allard C, Carneiro L, Fioramonti X, Collins S, Pénicaud L. Glucose and hypothalamic astrocytes: more than a fueling role? Neuroscience. 2016;323:110‐120. [DOI] [PubMed] [Google Scholar]
  • 234. Diano S, Kalra SP, Horvath TL. Leptin receptor immunoreactivity is associated with the Golgi apparatus of hypothalamic neurons and glial cells. J Neuroendocrinol. 1998;10(9):647‐650. [DOI] [PubMed] [Google Scholar]
  • 235. Pan W, Hsuchou H, He Y, et al. Astrocyte leptin receptor (ObR) and leptin transport in adult-onset obese mice. Endocrinology. 2008;149(6):2798‐2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Fuente-Martín E, García-Cáceres C, Argente-Arizón P, et al. Ghrelin regulates glucose and glutamate transporters in hypothalamic astrocytes. Sci Rep. 2016;6(1):23673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. García-Cáceres C, Quarta C, Varela L, et al. Astrocytic insulin signaling couples brain glucose uptake with nutrient availability. Cell. 2016;166(4):867‐880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Rottkamp DM, Rudenko IA, Maier MT, et al. Leptin potentiates astrogenesis in the developing hypothalamus. Mol Metab. 2015;4(11):881‐889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Kelley L, Verlezza S, Long H, Loka M, Walker C-D. Increased hypothalamic projections to the lateral hypothalamus and responses to leptin in rat neonates from high fat fed mothers. Front Neurosci. 2019;13:1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Park S, Jang A, Bouret SG. Maternal obesity-induced endoplasmic reticulum stress causes metabolic alterations and abnormal hypothalamic development in the offspring. PLoS Biol. 2020;18(3):e3000296‐e3000296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Dearden L, Buller S, Furigo IC, Fernandez-Twinn DS, Ozanne SE. Maternal obesity causes fetal hypothalamic insulin resistance and disrupts development of hypothalamic feeding pathways. Mol Metab. 2020;42:101079‐101079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Roberts BL, Bennett CM, Carroll JM, Lindsley SR, Kievit P. Early overnutrition alters synaptic signaling and induces leptin resistance in arcuate proopiomelanocortin neurons. Physiol Behav. 2019;206:166‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Lee CH, Song DK, Park CB, et al. Primary cilia mediate early life programming of adiposity through lysosomal regulation in the developing mouse hypothalamus. Nat Commun. 2020;11(1):5772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Reiter JF, Leroux MR. Genes and molecular pathways underpinning ciliopathies. Nat Rev Mol Cell Biol. 2017;18(9):533‐547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Stratigopoulos G, Martin Carli J, O’Day D, et al. Hypomorphism for RPGRIP1L, a ciliary gene vicinal to the FTO locus, causes increased adiposity in mice. Cell Metab. 2014;19(5):767‐779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Kang SS, Kurti A, Fair DA, Fryer JD. Dietary intervention rescues maternal obesity induced behavior deficits and neuroinflammation in offspring. J Neuroinflammation. 2014;11(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Rosin JM, Vora SR, Kurrasch DM. Depletion of embryonic microglia using the CSF1R inhibitor PLX5622 has adverse sex-specific effects on mice, including accelerated weight gain, hyperactivity and anxiolytic-like behaviour. Brain Behav Immun. 2018;73:682‐697. [DOI] [PubMed] [Google Scholar]
  • 248. Attig L, Solomon G, Ferezou J, et al. Early postnatal leptin blockage leads to a long-term leptin resistance and susceptibility to diet-induced obesity in rats. Int J Obes (Lond). 2008;32(7):1153‐1160. [DOI] [PubMed] [Google Scholar]
  • 249. Picó C, Oliver P, Sánchez J, et al. The intake of physiological doses of leptin during lactation in rats prevents obesity in later life. Int J Obes (Lond). 2007;31(8):1199‐1209. [DOI] [PubMed] [Google Scholar]
  • 250. Benoit C, Ould-Hamouda H, Crepin D, Gertler A, Amar L, Taouis M. Early leptin blockade predisposes fat-fed rats to overweight and modifies hypothalamic microRNAs. J Endocrinol. 2013;218(1):35‐47. [DOI] [PubMed] [Google Scholar]
  • 251. Plagemann A, Heidrich I, Götz F, Rohde W, Dörner G. Lifelong enhanced diabetes susceptibility and obesity after temporary intrahypothalamic hyperinsulinism during brain organization. Exp Clin Endocrinol. 1992;99(2):91‐95. [DOI] [PubMed] [Google Scholar]
  • 252. Steculorum SM, Collden G, Coupe B, et al. Neonatal ghrelin programs development of hypothalamic feeding circuits. J Clin Invest. 2015;125(2):846‐858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Johnson MD, Bouret SG, Dunn-Meynell AA, Boyle CN, Lutz TA, Levin BE. Early postnatal amylin treatment enhances hypothalamic leptin signaling and neural development in the selectively bred diet-induced obese rat. Am J Physiol Regul Integr Comp Physiol. 2016;311(6):R1032‐R1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Li C, Xu J-J, Hu H-T, et al. Amylin receptor insensitivity impairs hypothalamic POMC neuron differentiation in the male offspring of maternal high-fat diet-fed mice. Mol Metab. 2021;44:101135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Stoffers DA, Desai BM, DeLeon DD, Simmons RA. Neonatal exendin-4 prevents the development of diabetes in the intrauterine growth retarded rat. Diabetes. 2003;52(3):734‐740. [DOI] [PubMed] [Google Scholar]
  • 256. Rozo AV, Babu DA, Suen PA, et al. Neonatal GLP1R activation limits adult adiposity by durably altering hypothalamic architecture. Mol Metab. 2017;6(7):748‐759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Aoki N, Kawamura M, Matsuda T. Lactation-dependent down regulation of leptin production in mouse mammary gland. Biochimica et Biophysica Acta (BBA)—General Subjects. 1999;1427(2):298‐306. [DOI] [PubMed] [Google Scholar]
  • 258. Monks J, Orlicky DJ, Stefanski AL, et al. Maternal obesity during lactation may protect offspring from high fat diet-induced metabolic dysfunction. Nutr Diabetes. 2018;8(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Pomar CA, Castillo P, Palou M, Palou A, Picó C. Implementation of a healthy diet to lactating rats attenuates the early detrimental programming effects in the offspring born to obese dams. Putative relationship with milk hormone levels. J Nutr Biochem. 2022;107:109043. [DOI] [PubMed] [Google Scholar]
  • 260. Nozhenko Y, Asnani-Kishnani M, Rodríguez AM, Palou A. Milk leptin surge and biological rhythms of leptin and other regulatory proteins in breastmilk. PLoS One. 2015;10(12):e0145376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Zambrano E, Rodríguez-González GL, Reyes-Castro LA, et al. DHA Supplementation of obese rats throughout pregnancy and lactation modifies milk composition and anxiety behavior of offspring. Nutrients. 2021;13(12):4243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Casabiell X, Piñeiro V, Tomé MA, Peinó R, Dieguez C, Casanueva FF. Presence of leptin in colostrum and/or breast milk from lactating mothers: a potential role in the regulation of neonatal food intake. J Clin Endocrinol Metab. 1997;82(12):4270‐4273. [DOI] [PubMed] [Google Scholar]
  • 263. Sánchez J, Oliver P, Miralles O, Ceresi E, Picó C, Palou A. Leptin orally supplied to neonate rats is directly uptaken by the immature stomach and may regulate short-term feeding. Endocrinology. 2005;146(6):2575‐2582. [DOI] [PubMed] [Google Scholar]
  • 264. Sánchez J, Priego T, Palou M, Tobaruela A, Palou A, Picó C. Oral supplementation with physiological doses of leptin during lactation in rats improves insulin sensitivity and affects food preferences later in life. Endocrinology. 2008;149(2):733‐740. [DOI] [PubMed] [Google Scholar]
  • 265. Plagemann A, Harder T, Rake A, et al. Morphological alterations of hypothalamic nuclei due to intrahypothalamic hyperinsulinism in newborn rats. Int J Dev Neurosci. 1999;17(1):37‐44. [DOI] [PubMed] [Google Scholar]
  • 266. Harder T, Plagemann A, Rohde W, Dörner G. Syndrome X-like alterations in adult female rats due to neonatal insulin treatment. Metab Clin Exp. 1998;47(7):855‐862. [DOI] [PubMed] [Google Scholar]
  • 267. Kojima M, Hosoda H, et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656‐660. [DOI] [PubMed] [Google Scholar]
  • 268. Asakawa A, Inui A, Kaga T, et al. A role of ghrelin in neuroendocrine and behavioral responses to stress in mice. Neuroendocrinology. 2001;74(3):143‐147. [DOI] [PubMed] [Google Scholar]
  • 269. Zigman JM, Jones JE, Lee CE, Saper CB, Elmquist JK. Expression of ghrelin receptor mRNA in the rat and the mouse brain. J Comp Neurol. 2006;494(3):528‐548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Williams KW, Elmquist JK. From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat Neurosci. 2012;15(10):1350‐1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Tong J, D'Alessio D. Ghrelin and hypothalamic development: too little and too much of a good thing. J Clin Invest. 2015;125(2):490‐492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Collden G, Balland E, Parkash J, et al. Neonatal overnutrition causes early alterations in the central response to peripheral ghrelin. Mol Metab. 2015;4(1):15‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Ogawa A, Harris V, McCorkle SK, Unger RH, Luskey KL. Amylin secretion from the rat pancreas and its selective loss after streptozotocin treatment. J Clin Invest. 1990;85(3):973‐976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Sanke T, Hanabusa T, Nakano Y, et al. Plasma islet amyloid polypeptide (amylin) levels and their responses to oral glucose in type 2 (non-insulin-dependent) diabetic patients. Diabetologia. 1991;34(2):129‐132. [DOI] [PubMed] [Google Scholar]
  • 275. Mitsukawa T, Takemura J, Asai J, et al. Islet amyloid polypeptide response to glucose, insulin, and somatostatin analogue administration. Diabetes. 1990;39(5):639‐642. [DOI] [PubMed] [Google Scholar]
  • 276. Butler PC, Chou J, Carter WB, et al. Effects of meal ingestion on plasma amylin concentration in NIDDM and nondiabetic humans. Diabetes. 1990;39(6):752‐756. [DOI] [PubMed] [Google Scholar]
  • 277. Gedulin BR, Rink TJ, Young AA. Dose-response for glucagonostatic effect of amylin in rats. Metab Clin Exp. 1997;46(1):67‐70. [DOI] [PubMed] [Google Scholar]
  • 278. Qi T, Christopoulos G, Bailey RJ, Christopoulos A, Sexton PM, Hay DL. Identification of N-terminal receptor activity-modifying protein residues important for calcitonin gene-related peptide, adrenomedullin, and amylin receptor function. Mol Pharmacol. 2008;74(4):1059‐1071. [DOI] [PubMed] [Google Scholar]
  • 279. Le Foll C, Johnson MD, Dunn-Meynell AA, Boyle CN, Lutz TA, Levin BE. Amylin-induced central IL-6 production enhances ventromedial hypothalamic leptin signaling. Diabetes. 2015;64(5):1621‐1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Dunn-Meynell AA, Le Foll C, Johnson MD, Lutz TA, Hayes MR, Levin BE. Endogenous VMH amylin signaling is required for full leptin signaling and protection from diet-induced obesity. Am J Physiol Regul Integr Comp Physiol. 2016;310(4):R355‐R365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Lutz TA, Coester B, Whiting L, et al. Amylin selectively signals onto POMC neurons in the arcuate nucleus of the hypothalamus. Diabetes. 2018;67(5):805‐817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Rindi G, Terenghi G, Westermark G, Westermark P, Moscoso G, Polak JM. Islet amyloid polypeptide in proliferating pancreatic B cells during development, hyperplasia, and neoplasia in humans and mice. Am J Pathol. 1991;138(6):1321‐1334. [PMC free article] [PubMed] [Google Scholar]
  • 283. Mulder H, Myrsén-Axcrona U, Gebre-Medhin S, Ekblad E, Sundler F. Expression of non-classical islet hormone-like peptides during the embryonic development of the pancreas. Microsc Res Tech. 1998;43(4):313‐321. [DOI] [PubMed] [Google Scholar]
  • 284. Lutz TA, Le Foll C. Endogenous amylin contributes to birth of microglial cells in arcuate nucleus of hypothalamus and area postrema during fetal development. Am J Physiol Regul Integr Comp Physiol. 2019;316(6):R791‐R801. [DOI] [PubMed] [Google Scholar]
  • 285. Bouret SG, Gorski JN, Patterson CM, Chen S, Levin BE, Simerly RB. Hypothalamic neural projections are permanently disrupted in diet-induced obese rats. Cell Metab. 2008;7(2):179‐185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Abegg K, Hermann A, Boyle CN, Bouret SG, Lutz TA, Riediger T. Involvement of amylin and leptin in the development of projections from the area postrema to the nucleus of the solitary tract. Front Endocrinol (Lausanne). 2017;8:324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Hiles RA, Bawdon RE, Petrella EM. Ex vivo human placental transfer of the peptides pramlintide and exenatide (synthetic exendin-4). Hum Exp Toxicol. 2003;22(12):623‐628. [DOI] [PubMed] [Google Scholar]
  • 288. Kanoski SE, Hayes MR, Skibicka KP. GLP-1 and weight loss: unraveling the diverse neural circuitry. Am J Physiol Regul Integr Comp Physiol. 2016;310(10):R885‐R895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Ham JN, Crutchlow MF, Desai BM, Simmons RA, Stoffers DA. Exendin-4 normalizes islet vascularity in intrauterine growth restricted rats: potential role of VEGF. Pediatr Res. 2009;66(1):42‐46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Rajamoorthi A, LeDuc CA, Thaker VV. The metabolic conditioning of obesity: A review of the pathogenesis of obesity and the epigenetic pathways that “program” obesity from conception. Front Endocrinol (Lausanne). 2022;13:1032491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Marco A, Kisliouk T, Tabachnik T, Meiri N, Weller A. Overweight and CpG methylation of the Pomc promoter in offspring of high-fat-diet-fed dams are not “reprogrammed” by regular chow diet in rats. FASEB J. 2014;28(9):4148‐4157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Shi X, Wang X, Li Q, et al. Nuclear factor kappaB (NF-kappaB) suppresses food intake and energy expenditure in mice by directly activating the Pomc promoter. Diabetologia. 2013;56(4):925‐936. [DOI] [PubMed] [Google Scholar]
  • 293. Plagemann A, Harder T, Brunn M, et al. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome. J Physiol. 2009;587(Pt 20):4963‐4976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. Gemma C, Sookoian S, Alvariñas J, et al. Maternal pregestational BMI is associated with methylation of the PPARGC1A promoter in newborns. Obesity (Silver Spring). 2009;17(5):1032‐1039. [DOI] [PubMed] [Google Scholar]
  • 295. Yang Q-Y, Liang J-F, Rogers CJ, Zhao J-X, Zhu M-J, Du M. Maternal obesity induces epigenetic modifications to facilitate Zfp423 expression and enhance adipogenic differentiation in fetal mice. Diabetes. 2013;62(11):3727‐3735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Borengasser SJ, Zhong Y, Kang P, et al. Maternal obesity enhances white adipose tissue differentiation and alters genome-scale DNA methylation in male rat offspring. Endocrinology. 2013;154(11):4113‐4125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Hjort L, Martino D, Grunnet LG, et al. Gestational diabetes and maternal obesity are associated with epigenome-wide methylation changes in children. JCI Insight. 2018;3(17):e122572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298. Gali Ramamoorthy T, Allen T-J, Davies A, et al. Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int J Obes (Lond). 2018;42(8):1431‐1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299. Schellong K, Melchior K, Ziska T, et al. Hypothalamic insulin receptor expression and DNA promoter methylation are sex-specifically altered in adult offspring of high-fat diet (HFD)-overfed mother rats. J Nutr Biochem. 2019;67:28‐35. [DOI] [PubMed] [Google Scholar]
  • 300. Sharp GC, Lawlor DA, Richmond RC, et al. Maternal pre-pregnancy BMI and gestational weight gain, offspring DNA methylation and later offspring adiposity: findings from the Avon Longitudinal Study of Parents and Children. Int J Epidemiol. 2015;44(4):1288‐1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Morales E, Groom A, Lawlor DA, Relton CL. DNA Methylation signatures in cord blood associated with maternal gestational weight gain: results from the ALSPAC cohort. BMC Res Notes. 2014;7(1):278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Liu X, Chen Q, Tsai H, et al. Maternal preconception body mass index and offspring cord blood DNA methylation: exploration of early life origins of disease. Environ Mol Mutagen. 2014;55(3):223‐230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Martin CL, Jima D, Sharp GC, et al. Maternal pre-pregnancy obesity, offspring cord blood DNA methylation, and offspring cardiometabolic health in early childhood: an epigenome-wide association study. Epigenetics. 2019;14(4):325‐340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Sharp GC, Salas LA, Monnereau C, et al. Maternal BMI at the start of pregnancy and offspring epigenome-wide DNA methylation: findings from the pregnancy and childhood epigenetics (PACE) consortium. Hum Mol Genet. 2017;26(20):4067‐4085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305. Vehmeijer FOL, Küpers LK, Sharp GC, et al. DNA Methylation and body mass index from birth to adolescence: meta-analyses of epigenome-wide association studies. Genome Med. 2020;12(1):105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. Berglind D, Müller P, Willmer M, et al. Differential methylation in inflammation and type 2 diabetes genes in siblings born before and after maternal bariatric surgery. Obesity (Silver Spring). 2016;24(1):250‐261. [DOI] [PubMed] [Google Scholar]
  • 307. Mitsuya K, Parker AN, Liu L, Ruan J, Vissers MCM, Myatt L. Alterations in the placental methylome with maternal obesity and evidence for metabolic regulation. PLOS ONE. 2017;12(10):e0186115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Lesseur C, Armstrong DA, Paquette AG, Li Z, Padbury JF, Marsit CJ, . Maternal obesity and gestational diabetes are associated with placental leptin DNA methylation. Am J Obstet Gynecol. 2014;211(6):654 e1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Nogues P, Dos Santos E, Jammes H, et al. Maternal obesity influences expression and DNA methylation of the adiponectin and leptin systems in human third-trimester placenta. Clin Epigenetics. 2019;11(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310. Bouchard L, Thibault S, Guay S-P, et al. Leptin gene epigenetic adaptation to impaired glucose metabolism during pregnancy. Diabetes Care. 2010;33(11):2436‐2441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. King SE, Skinner MK. Epigenetic transgenerational inheritance of obesity susceptibility. Trends Endocrinol Metab. 2020;31(7):478‐494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. Dunn GA, Bale TL. Maternal high-fat diet effects on third-generation female body size via the paternal lineage. Endocrinology. 2011;152(6):2228‐2236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Raad G, Serra F, Martin L, et al. Paternal multigenerational exposure to an obesogenic diet drives epigenetic predisposition to metabolic diseases in mice. Elife. 2021;10:e61736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Bernal AJ, Jirtle RL. Epigenomic disruption: the effects of early developmental exposures. Birth Defects Res A Clin Mol Teratol. 2010;88(10):938‐944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Waterland RA, Travisano M, Tahiliani KG, Rached MT, Mirza S. Methyl donor supplementation prevents transgenerational amplification of obesity. Int J Obes (Lond). 2008;32(9):1373‐1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. Jiao F, Yan X, Yu Y, et al. Protective effects of maternal methyl donor supplementation on adult offspring of high fat diet-fed dams. J Nutr Biochem. 2016;34:42‐51. [DOI] [PubMed] [Google Scholar]
  • 317. Vucetic Z, Kimmel J, Totoki K, Hollenbeck E, Reyes TM. Maternal high-fat diet alters methylation and gene expression of dopamine and opioid-related genes. Endocrinology. 2010;151(10):4756‐4764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Carlin J, George R, Reyes TM. Methyl donor supplementation blocks the adverse effects of maternal high fat diet on offspring physiology. PLoS One. 2013;8(5):e63549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319. Birney E, Smith GD, Greally JM. Epigenome-wide association studies and the interpretation of disease -omics. PLoS Genet. 2016;12(6):e1006105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Michels KB, Binder AM, Dedeurwaerder S, et al. Recommendations for the design and analysis of epigenome-wide association studies. Nat Methods. 2013;10(10):949‐955. [DOI] [PubMed] [Google Scholar]
  • 321. Semple BD, Blomgren K, Gimlin K, Ferriero DM, Noble-Haeusslein LJ. Brain development in rodents and humans: identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol. 2013;106–107:1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322. Kempermann G, Kuhn HG, Gage FH. More hippocampal neurons in adult mice living in an enriched environment. Nature. 1997;386(6624):493‐495. [DOI] [PubMed] [Google Scholar]
  • 323. Cameron HA, Woolley CS, McEwen BS, Gould E. Differentiation of newly born neurons and glia in the dentate gyrus of the adult rat. Neuroscience. 1993;56(2):337‐344. [DOI] [PubMed] [Google Scholar]
  • 324. Eriksson PS, Perfilieva E, Björk-Eriksson T, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313‐1317. [DOI] [PubMed] [Google Scholar]
  • 325. Shimogori T, Lee DA, Miranda-Angulo A, et al. A genomic atlas of mouse hypothalamic development. Nat Neurosci. 2010;13(6):767‐775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326. Zhou X, Lu Y, Zhao F, et al. Deciphering the spatial-temporal transcriptional landscape of human hypothalamus development. Cell Stem Cell. 2022;29(2):328‐343.e5. [DOI] [PubMed] [Google Scholar]
  • 327. Koutcherov Y, Mai JK, Ashwell KWS, Paxinos G. Organization of human hypothalamus in fetal development. J Comp Neurol. 2002;446(4):301‐324. [DOI] [PubMed] [Google Scholar]
  • 328. Giedd JN, Blumenthal J, Jeffries NO, et al. Brain development during childhood and adolescence: a longitudinal MRI study. Nat Neurosci. 1999;2(10):861‐863. [DOI] [PubMed] [Google Scholar]
  • 329. Paus T, Zijdenbos A, Worsley K, et al. Structural maturation of neural pathways in children and adolescents: in vivo study. Science. 1999;283(5409):1908‐1911. [DOI] [PubMed] [Google Scholar]
  • 330. Downes N, Mullins P. The development of myelin in the brain of the juvenile rat. Toxicol Pathol. 2014;42(5):913‐922. [DOI] [PubMed] [Google Scholar]
  • 331. Bjelke B, Seiger A. Morphological distribution of MBP-like immunoreactivity in the brain during development. Int J Dev Neurosci. 1989;7(2):145‐164. [DOI] [PubMed] [Google Scholar]
  • 332. Dobbing J, Sands J. Comparative aspects of the brain growth spurt. Early Hum Dev. 1979;3(1):79‐83. [DOI] [PubMed] [Google Scholar]
  • 333. Bockhorst KH, Narayana PA, Liu R, et al. Early postnatal development of rat brain: in vivo diffusion tensor imaging. J Neurosci Res. 2008;86(7):1520‐1528. [DOI] [PubMed] [Google Scholar]
  • 334. Baloch S, Verma R, Huang H, et al. Quantification of brain maturation and growth patterns in C57BL/6J mice via computational neuroanatomy of diffusion tensor images. Cereb Cortex. 2009;19(3):675‐687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Demerath EW, Johnson W, Davern BA, et al. New body composition reference charts for preterm infants. Am J Clin Nutr. 2017;105(1):70‐77. [DOI] [PubMed] [Google Scholar]
  • 336. Ziegler EE, O'Donnell AM, Nelson SE, Fomon SJ. Body composition of the reference fetus. Growth. 1976;40(4):329‐341. [PubMed] [Google Scholar]
  • 337. Hawkes CP, Hourihane JO, Kenny LC, Irvine AD, Kiely M, Murray DM. Gender- and gestational age–specific body fat percentage at birth. Pediatrics. 2011;128(3):e645. [DOI] [PubMed] [Google Scholar]
  • 338. Ellis KJ. Human body composition: in vivo methods. Physiol Rev. 2000;80(2):649‐680. [DOI] [PubMed] [Google Scholar]
  • 339. Hoggard N, Hunter L, Duncan JS, Williams LM, Trayhurn P, Mercer JG. Leptin and leptin receptor mRNA and protein expression in the murine fetus and placenta. Proc Natl Acad Sci U S A. 1997;94(20):11073‐11078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Hoggard N, Hunter L, Lea RG, Trayhurn P, Mercer JG. Ontogeny of the expression of leptin and its receptor in the murine fetus and placenta. Br J Nutr. 2000;83(3):317‐326. [DOI] [PubMed] [Google Scholar]
  • 341. Henson MC, Castracane VD, O’Neil JS, et al. Serum leptin concentrations and expression of leptin transcripts in placental trophoblast with advancing baboon pregnancy. J Clin Endocrinol Metab. 1999;84(7):2543‐2549. [DOI] [PubMed] [Google Scholar]
  • 342. Butte NF, Hopkinson JM, Nicolson MA. Leptin in human reproduction: serum leptin levels in pregnant and lactating women. J Clin Endocrinol Metab. 1997;82(2):585‐589. [DOI] [PubMed] [Google Scholar]
  • 343. Hardie L, Trayhurn P, Abramovich D, Fowler P. Circulating leptin in women: a longitudinal study in the menstrual cycle and during pregnancy. Clin Endocrinol (Oxf). 1997;47(1):101‐106. [DOI] [PubMed] [Google Scholar]
  • 344. Highman TJ, Friedman JE, Huston LP, Wong WW, Catalano PM. Longitudinal changes in maternal serum leptin concentrations, body composition, and resting metabolic rate in pregnancy. Am J Obstet Gynecol. 1998;178(5):1010‐1015. [DOI] [PubMed] [Google Scholar]
  • 345. Sattar N, Greer IA, Pirwani I, Gibson J, Wallace AM. Leptin levels in pregnancy: marker for fat accumulation and mobilization? Acta Obstet Gynecol Scand. 1998;77(3):278‐283. [PubMed] [Google Scholar]
  • 346. Henson MC, Castracane VD. Leptin in pregnancy. Biol Reprod. 2000;63(5):1219‐1228. [DOI] [PubMed] [Google Scholar]
  • 347. Lage M, Garcia-Mayor RV, Tomé MA, et al. Serum leptin levels in women throughout pregnancy and the postpartum period and in women suffering spontaneous abortion. Clin Endocrinol (Oxf). 1999;50(2):211‐216. [DOI] [PubMed] [Google Scholar]
  • 348. Sivan E, Whittaker PG, Sinha D, et al. Leptin in human pregnancy: the relationship with gestational hormones. Am J Obstet Gynecol. 1998;179(5):1128‐1132. [DOI] [PubMed] [Google Scholar]
  • 349. Misra VK, Trudeau S. The influence of overweight and obesity on longitudinal trends in maternal serum leptin levels during pregnancy. Obesity (Silver Spring). 2011;19(2):416‐421. [DOI] [PubMed] [Google Scholar]
  • 350. Misra VK, Straughen JK, Trudeau S. Maternal serum leptin during pregnancy and infant birth weight: the influence of maternal overweight and obesity. Obesity (Silver Spring). 2013;21(5):1064‐1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351. Castellano Filho DS, Correa JOA, Ramos PS, Montessi MO, Aarestrup BJV, Aarestrup FM. Body weight gain and serum leptin levels of non-overweight and overweight/obese pregnant women. Med Sci Monit. 2013;19:1043‐1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352. Cetin I, Morpurgo PS, Radaelli T, et al. Fetal plasma leptin concentrations: relationship with different intrauterine growth patterns from 19 weeks to term. Pediatr Res. 2000;48(5):646‐651. [DOI] [PubMed] [Google Scholar]
  • 353. Karakosta P, Chatzi L, Plana E, Margioris A, Castanas E, Kogevinas M. Leptin levels in cord blood and anthropometric measures at birth: a systematic review and meta-analysis. Paediatr Perinat Epidemiol. 2011;25(2):150‐163. [DOI] [PubMed] [Google Scholar]
  • 354. Kawamata R, Suzuki Y, Yada Y, et al. Gut hormone profiles in preterm and term infants during the first 2 months of life. J Pediatr Endocrinol Metab. 2014;27(7-8):717‐723. [DOI] [PubMed] [Google Scholar]
  • 355. Jaquet D, Leger J, Levy-Marchal C, Oury JF, Czernichow P. Ontogeny of leptin in human fetuses and newborns: effect of intrauterine growth retardation on serum leptin concentrations. J Clin Endocrinol Metab. 1998;83(4):1243‐1246. [DOI] [PubMed] [Google Scholar]
  • 356. Yildiz L, Avci B, Ingeç M. Umbilical cord and maternal blood leptin concentrations in intrauterine growth retardation. Clin Chem Lab Med. 2002;40(11):1114‐1117. [DOI] [PubMed] [Google Scholar]
  • 357. Valūnienė M, Verkauskienė R, Boguszewski M, et al. Leptin levels at birth and in early postnatal life in small- and appropriate-for-gestational-age infants. Medicina (Kaunas). 2007;43(10):784‐791. [PubMed] [Google Scholar]
  • 358. Koistinen HA, Koivisto VA, Andersson S, et al. Leptin concentration in cord blood correlates with intrauterine growth*. The Journal of Clinical Endocrinology & Metabolism. 1997;82(10):3328‐3330. [DOI] [PubMed] [Google Scholar]
  • 359. Marchini G, Fried G, Östlund E, Hagenäs L. Plasma leptin in infants: relations to birth weight and weight loss. Pediatrics. 1998;101(3):429‐432. [DOI] [PubMed] [Google Scholar]
  • 360. Taricco E, Radaelli T, Nobile de Santis MS, Cetin I. Foetal and placental weights in relation to maternal characteristics in gestational diabetes. Placenta. 2003;24(4):343‐347. [DOI] [PubMed] [Google Scholar]
  • 361. Ertl T, Funke S, Sárkány I, et al. Postnatal changes of leptin levels in full-term and preterm neonates: their relation to intrauterine growth, gender and testosterone. Biol Neonate. 1999;75(3):167‐176. [DOI] [PubMed] [Google Scholar]
  • 362. Gómez L, Carrascosa A, Yeste D, et al. Leptin values in placental cord blood of human newborns with normal intrauterine growth after 30–42 weeks of gestation. Horm Res. 1999;51(1):10‐14. [DOI] [PubMed] [Google Scholar]
  • 363. Matsuda J, Yokota I, Iida M, et al. Serum leptin concentration in cord blood: relationship to birth weight and gender. J Clin Endocrinol & Metabol. 1997;82(5):1642‐1644. [DOI] [PubMed] [Google Scholar]
  • 364. Helland IB, Reseland JE, Saugstad OD, Drevon CA. Leptin levels in pregnant women and newborn infants: gender differences and reduction during the neonatal period. Pediatrics. 1998;101(3):E12. [DOI] [PubMed] [Google Scholar]
  • 365. Fomon SJ, Haschke F, Ziegler EE, Nelson SE. Body composition of reference children from birth to age 10 years. Am J Clin Nutr. 1982;35(5):1169‐1175. [DOI] [PubMed] [Google Scholar]
  • 366. Butte NF, Hopkinson JM, Wong WW, Smith EO, Ellis KJ. Body composition during the first 2 years of life: an updated reference. Pediatr Res. 2000;47(5):578‐585. [DOI] [PubMed] [Google Scholar]
  • 367. HorlickK MB, Rosenbaum M, Nicholson M, et al. Effect of puberty on the relationship between circulating leptin and body composition. J Clin Endocrinol Metab. 2000;85(7):2509‐2518. [DOI] [PubMed] [Google Scholar]
  • 368. Ellis KJ, Nicolson M. Leptin levels and body fatness in children: effects of gender, ethnicity, and sexual development. Pediatr Res. 1997;42(4):484‐488. [DOI] [PubMed] [Google Scholar]
  • 369. Blum WF, Englaro P, Hanitsch S, et al. Plasma leptin levels in healthy children and adolescents: dependence on body mass index, body fat mass, gender, pubertal stage, and testosterone. J Clin Endocrinol Metab. 1997;82(9):2904‐2910. [DOI] [PubMed] [Google Scholar]
  • 370. Garcia-Mayor RV, Andrade MA, Rios M, Lage M, Dieguez C, Casanueva FF. Serum leptin levels in normal children: relationship to age, gender, body mass index, pituitary-gonadal hormones, and pubertal stage. J Clin Endocrinol Metab. 1997;82(9):2849‐2855. [DOI] [PubMed] [Google Scholar]
  • 371. Nagy TR, Gower BA, Trowbridge CA, Dezenberg C, Shewchuk RM, Goran MI. Effects of gender, ethnicity, body composition, and fat distribution on serum leptin concentrations in children. J Clin Endocrinol Metab. 1997;82(7):2148‐2152. [DOI] [PubMed] [Google Scholar]
  • 372. Clayton PE, Gill MS, Hall CM, Tillmann V, Whatmore AJ, Price DA. Serum leptin through childhood and adolescence. Clin Endocrinol (Oxf). 1997;46(6):727‐733. [DOI] [PubMed] [Google Scholar]
  • 373. Rosenbaum M, Nicolson M, Hirsch J, et al. Effects of gender, body composition, and menopause on plasma concentrations of leptin. J Clin Endocrinol Metab. 1996;81(9):3424‐3427. [DOI] [PubMed] [Google Scholar]
  • 374. Rosenbaum M, Pietrobelli A, Vasselli JR, Heymsfield SB, Leibel RL. Sexual dimorphism in circulating leptin concentrations is not accounted for by differences in adipose tissue distribution. Int J Obes Relat Metab Disord. 2001;25(9):1365‐1371. [DOI] [PubMed] [Google Scholar]
  • 375. Kennedy A, Gettys TW, Watson P, et al. The metabolic significance of leptin in humans: gender-based differences in relationship to adiposity, insulin sensitivity, and energy expenditure. J Clin Endocrinol Metab. 1997;82(4):1293‐1300. [DOI] [PubMed] [Google Scholar]
  • 376. Linnemann K, Malek A, Sager R, Blum WF, Schneider H, Fusch C. Leptin production and release in the dually in vitro perfused human placenta. J Clin Endocrinol Metab. 2000;85(11):4298‐4301. [DOI] [PubMed] [Google Scholar]
  • 377. Lepercq J, Challier J-C, Guerre-Millo M, Cauzac M, Vidal H, Hauguel-de Mouzon S. Prenatal leptin production: evidence that fetal adipose tissue produces leptin. J Clin Endocrinol Metab. 2001;86(6):2409‐2413. [DOI] [PubMed] [Google Scholar]
  • 378. Hoggard N, Crabtree J, Allstaff S, Abramovich DR, Haggarty P. Leptin secretion to both the maternal and fetal circulation in the ex vivo perfused human term placenta. Placenta. 2001;22(4):347‐352. [DOI] [PubMed] [Google Scholar]
  • 379. Hecht JL, Kliman HJ, Allred EN, et al. Reference weights for placentas delivered before the 28th week of gestation. Placenta. 2007;28(10):987‐990. [DOI] [PubMed] [Google Scholar]
  • 380. Burkhardt T, Schäffer L, Schneider C, Zimmermann R, Kurmanavicius J. Reference values for the weight of freshly delivered term placentas and for placental weight-birth weight ratios. Eur J Obstet Gynecol Reprod Biol. 2006;128(1-2):248‐252. [DOI] [PubMed] [Google Scholar]
  • 381. Hassink SG, de Lancey E, Sheslow DV, et al. Placental leptin: an important new growth factor in intrauterine and neonatal development? Pediatrics. 1997;100(1):E1. [DOI] [PubMed] [Google Scholar]
  • 382. Gavrilova O, Barr V, Marcus-Samuels B, Reitman M. Hyperleptinemia of pregnancy associated with the appearance of a circulating form of the leptin receptor. J Biol Chem. 1997;272(48):30546‐30551. [DOI] [PubMed] [Google Scholar]
  • 383. Smith JT, Waddell BJ. Leptin distribution and metabolism in the pregnant rat: transplacental leptin passage increases in late gestation but is reduced by excess glucocorticoids. Endocrinology. 2003;144(7):3024‐3030. [DOI] [PubMed] [Google Scholar]
  • 384. Malik NM, Carter ND, Wilson CA, Scaramuzzi RJ, Stock MJ, Murray JF. Leptin expression in the fetus and placenta during mouse pregnancy. Placenta. 2005;26(1):47‐52. [DOI] [PubMed] [Google Scholar]
  • 385. McCurdy CE, Bishop JM, Williams SM, et al. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J Clin Invest. 2009;119(2):323‐335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386. Schubring C, Prohaska F, Prohaska A, et al. Leptin concentrations in maternal serum and amniotic fluid during the second trimenon: differential relation to fetal gender and maternal morphometry. Eur J Obstet Gynecol Reprod Biol. 1999;86(2):151‐157. [DOI] [PubMed] [Google Scholar]
  • 387. Hytinantti T, Koistinen HA, Koivisto VA, Karonen S-L, Andersson S. Changes in leptin concentration during the early postnatal period: adjustment to extrauterine life? Pediatr Res. 1999;45(2):197‐201. [DOI] [PubMed] [Google Scholar]
  • 388. Long NM, Ford SP, Nathanielsz PW. Maternal obesity eliminates the neonatal lamb plasma leptin peak. J Physiol. 2011;589(Pt 6):1455‐1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389. Smith AM, Pankey CL, Odhiambo JF, Ghnenis AB, Nathanielsz PW, Ford SP. Rapid communication: reduced maternal nutrition during early- to mid-gestation elevates newborn lamb plasma cortisol concentrations and eliminates the neonatal leptin surge. J Anim Sci. 2018;96(7):2640‐2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390. Bispham J, Budge H, Mostyn A, et al. Ambient temperature, maternal dexamethasone, and postnatal ontogeny of leptin in the neonatal lamb. Pediatr Res. 2002;52(1):85‐90. [DOI] [PubMed] [Google Scholar]
  • 391. Long NM, George LA, Uthlaut AB, et al. Maternal obesity and increased nutrient intake before and during gestation in the ewe results in altered growth, adiposity, and glucose tolerance in adult offspring. J Anim Sci. 2010;88(11):3546‐3553. [DOI] [PubMed] [Google Scholar]
  • 392. Ford SP, Hess BW, Schwope MM, et al. Maternal undernutrition during early to mid-gestation in the ewe results in altered growth, adiposity, and glucose tolerance in male offspring. J Anim Sci. 2007;85(5):1285‐1294. [DOI] [PubMed] [Google Scholar]
  • 393. Grayson BE, Allen SE, Billes SK, Williams SM, Smith MS, Grove KL. Prenatal development of hypothalamic neuropeptide systems in the nonhuman primate. Neuroscience. 2006;143(4):975‐986. [DOI] [PubMed] [Google Scholar]
  • 394. Dodd JM, Turnbull D, McPhee AJ, et al. Antenatal lifestyle advice for women who are overweight or obese: LIMIT randomised trial. BMJ. 2014;348(feb10 3):g1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395. Grivell RM, Yelland LN, Deussen A, Crowther CA, Dodd JM. Antenatal dietary and lifestyle advice for women who are overweight or obese and the effect on fetal growth and adiposity: the LIMIT randomised trial. BJOG: An International Journal of Obstetrics & Gynaecology. 2016;123(2):233‐243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396. Oteng-Ntim E, Varma R, Croker H, Poston L, Doyle P. Lifestyle interventions for overweight and obese pregnant women to improve pregnancy outcome: systematic review and meta-analysis. BMC Med. 2012;10(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Endocrine Reviews are provided here courtesy of The Endocrine Society

RESOURCES