Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2019

Open Access 01.12.2019 | Review

Effects of high-fat diets on fetal growth in rodents: a systematic review

verfasst von: Julian K. Christians, Kendra I. Lennie, Lisa K. Wild, Raajan Garcha

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2019

Abstract

Background

Maternal nutrition during pregnancy has life-long consequences for offspring. However, the effects of maternal overnutrition and/ or obesity on fetal growth remain poorly understood, e.g., it is not clear why birthweight is increased in some obese pregnancies but not in others. Maternal obesity is frequently studied using rodents on high-fat diets, but effects on fetal growth are inconsistent. The purpose of this review is to identify factors that contribute to reduced or increased fetal growth in rodent models of maternal overnutrition.

Methods

We searched Web of Science and screened 2173 abstracts and 328 full texts for studies that fed mice or rats diets providing ~ 45% or ~ 60% calories from fat for 3 weeks or more prior to pregnancy. We identified 36 papers matching the search criteria that reported birthweight or fetal weight.

Results

Studies that fed 45% fat diets to mice or 60% fat diets to rats generally did not show effects on fetal growth. Feeding a 45% fat diet to rats generally reduced birth and fetal weight. Feeding mice a 60% fat diet for 4–9 weeks prior to pregnancy tended to increase in fetal growth, whereas feeding this diet for a longer period tended to reduce fetal growth.

Conclusions

The high-fat diets used most often with rodents do not closely match Western diets and frequently reduce fetal growth, which is not a typical feature of obese human pregnancies. Adoption of standard protocols that more accurately mimic effects on fetal growth observed in obese human pregnancies will improve translational impact in this field.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12958-019-0482-y) contains supplementary material, which is available to authorized users.
Abkürzungen
BMI
Body mass index
GDM
Gestational diabetes mellitus
HFD
High fat diet
LGA
Large-for-gestational-age

Background

The prenatal environment has far reaching effects on health throughout life [1]. Among the first observations of such effects were associations between birthweight and risk of cardiovascular disease and diabetes [25]. Initial studies found associations between low birthweight and adverse health outcomes later in life, whereas subsequent studies found that both very low and very high birthweight increased the risk of adult disease [68]. As a result of these associations, there is enormous interest in the long-term consequences of maternal overnutrition and/ or obesity during pregnancy [9]. However, the effects of maternal obesity on birthweight remain poorly understood.
Studies consistently report that obese women are at higher risk of having a large-for-gestational-age (LGA) baby [6, 10]. A higher frequency of LGA is due in part to obesity increasing the risk of gestational diabetes mellitus (GDM), which increases the risk of LGA [11]. However, maternal obesity also increases the risk of LGA in the absence of GDM [12]. Obese women are also at higher risk of hypertensive disorders of pregnancy such as preeclampsia, which is often associated with small-for-gestational-age (SGA) neonates [11]. This observation suggests that obese mothers might be at higher risk of SGA as well, but such an association is not observed consistently [13, 14]. There have been numerous reviews of the effects of dietary, exercise and lifestyle interventions on birthweight [13, 1518], as well as associations between dietary intake and birthweight [13, 1921]. However, these have not addressed why fetal growth is normal in many obese and diabetic pregnancies but altered in others [22].
Animal models are needed to better understand the effects of maternal obesity on fetal growth, and indeed numerous such studies have been performed, many using rodents fed high-fat diets (HFD). However, there is little standardization in this field, and the human phenotype being modeled is rarely defined more specifically than “obesity in pregnancy”. While there are numerous reviews of the effects of maternal overnutrition and/ or obesity on offspring glycemic control [23] and cardiovascular health [24, 25] in animal models, to our knowledge there has been no review of the effects of maternal HFD on birthweight in rodents. Rodents are born at a different developmental stage than humans, with birth in rodents corresponding to the end of the second trimester/ beginning of the third trimester in humans [26]. Nevertheless, hundreds of studies use rodent pregnancy as a model of human pregnancy, and thus it is necessary to consider to what extent rodent models mimic humans with respect to fetal growth; rodent birthweight is expected to provide a model of human fetal growth over the first two trimesters.
The purpose of this review is to identify factors (macronutrient composition, duration of diet, strain, etc.) that contribute to reduced or increased fetal growth in rodent models of maternal overnutrition. Such a review is needed to enable the development of more relevant and standardized animal models of human phenotypes. We sought to identify factors that are common among animal models that report an increase or decrease in birthweight, and to assess the uniformity of studies that use similar models. Our aim was not to perform a meta-analysis to assess whether, on average, there was an effect of HFD on birthweight (e.g., [27]). Rather, our goal was to review studies with different methodologies to determine whether some manipulations yielded consistent effects on birthweight.

Methods

We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [28].
The Web of Science database was searched using the terms: (maternal OR gestational) AND diet AND (birth OR fetal) AND weight AND (mouse OR rat). An initial search was conducted on July 12th, 2016, and an updated search was conducted on July 4, 2018 (the latter including only studies from 2016 to 2018). Figure 1 shows the selection process.

Eligibility criteria

The participants, interventions, comparisons, outcomes, and study design (PICOS) criteria were as follows: participants were mice or rats; intervention was ad libitum access to a HFD during pregnancy (experimental diet may have also been provided prior to pregnancy, or may have been provided for only part of pregnancy); comparison was with a control diet provided ad libitum; outcome was birthweight (postnatal day 2 or earlier) or fetal weight of offspring from manipulated pregnancies (i.e., effects on subsequent generations were not included); study design was a controlled experiment. We included studies with any type of control diet that had a lower fat content than the intervention. Only studies published in English were included. Where more than one study appeared to describe the same dataset, only the first publication was included. Our updated, refined search added the following criteria: the HFD was provided for 3 weeks or more prior to pregnancy, and was either 44–45% calories from fat, or 57–62% calories from fat; cafeteria diets where animals were able to choose among food items were excluded.

Study selection, data items and summary measures

In the initial search, 1954 titles and abstracts were screened and the full texts of 275 papers were reviewed. 83 studies reporting the birth or fetal weight of offspring were identified (Fig. 1). From each of these, the following variables were recorded: percentage fat content in the HFD, source of additional fat in the HFD (plant vs. animal), duration of diet prior to pregnancy, sample size per group, and age of dams at mating. Fat contents expressed as percentage of calories from fat were recorded if available. Our summary measure was whether the maternal HFD led to a statistically significant increase or decrease in birth and/ or fetal weight.
In the updated search using refined criteria, 219 titles and abstracts were screened, and after removing studies already identified in the initial search, 53 full texts were reviewed. This yielded 11 new papers reporting birth or fetal weight of offspring and, together with studies from the initial search, 36 papers were identified that matched the refined criteria (Fig. 1). In addition to the information collected for the initial search, we also recorded how maternal weight was affected by the experimental diet, whether maternal glucose tolerance was affected, whether litter size was affected, and more information regarding the control diet.
We assumed that animals were assigned to experimental groups at random, and therefore that there was little risk of bias within individual studies. However, we acknowledge that there was likely a publication bias across studies, whereby statistically significant effects were more likely to be reported.

Results

The studies identified in the initial search, and the data extracted from each, are provided in Additional file 1: Table S1. Table 1 shows the number of studies finding an increase, decrease, or no effect on birthweight, aggregated by species, fat content, and duration of diet. More studies used rat than mouse as a model organism, and most used a HFD where the percentage of calories from fat was 45% or greater and included fat from animal sources (often lard) (Table 1, Additional file 1: Table S1). Among these studies, there were no consistent patterns with respect to effects on fetal growth. Increases and decreases were seen in studies of rats and mice, regardless of fat content, fat source or duration of diet prior to pregnancy. Although it might be expected that significant effects on birthweight would be found more often in studies with larger sample sizes, significant effects were approximately as frequent in studies with small sample sizes (< 10 dams per group) as in studies with moderate sample sizes (10–20 per group); few studies had greater than 20 dams per group (Additional file 1: Table S1).
Table 1
Summary of studies identified in initial search
Experimental protocol
Effect on fetal growth
Calories from fat
Total
<  45%
45%
>  45%
Cafeteria
Rats fed high-fat diet for 3 weeks or more prior to pregnancy
Decrease
0
4
2
4
10
  
Nivoit et al. (2009) [29]; Hayes et al. (2012) [30]; Christante et al. (2013)b [31]; Reynolds et al. (2014) [32]
Couvreur et al. (2011) [33]; Li et al. (2012)b [34]
Akyol et al. (2009) [35]; Raipuria et al. (2015) [36]; Ramirez-Lopez et al. (2016) [37]; Sánchez-Blanco et al. (2016) [38]
 
No effect
3
2
8
3
16
 
Del Prado et al. (1997) [39]; Caluwaerts et al. 2007 [40]; Nitert et al. (2013) [41]
Zambrano et al. (2010)b [42]; Vega et al. (2015) b [43]
Shaw et al. (1997)b [44]; Srinivasan et al. (2006) [45]; Ferezou-Viala et al. (2007) [46]; Gupta et al. (2009) [47]; Mitra et al. (2009) [48]; White et al. (2009) [49]; Guberman et al. (2013) [50]; Desai et al. (2014) [51]
Rolls and Rowe (1982) [52]; Chen et al. (2008) [53]; Ong and Muhlhausler (2011) [54]
 
Increase
0
1
2
1
4
  
Song et al. (2015) [55]
Gaccioli et al. (2013)b [56]; Mazzucco et al. (2013) [57]
Yang et al. (2015) [58]
 
Rats fed high-fat diet for less than 3 weeks prior to pregnancy
Decrease
1
5
0
3
9
 
Cerf et al. (2005) [59]
Mark et al. (2011) [60]; Smith et al. (2014) [61]; Cordero et al. (2015) [62]; Cunha et al. (2015)b [63]; Segovia et al. (2015) [64]
 
Bayol et al. (2007) [65]; Zhang et al. (2011) [66]; Gugusheff et al. (2013) [67]
 
No effect
2
2
4
2
10
 
Martin et al. (1987) [68]; Cerf et al. (2010)a [69]
Yang et al. (2012) [70]; Tsoulis et al. (2016) [71]
Ebesh et al. (1999) [72]; Walker et al. (2008)b [73]; Lin et al. (2011)b [74]; de Oliveira Andrade et al. (2014) [75]
Rolls et al. (1984) [76]; Zhang et al. (2013) [77]
 
Increase
4
1
1
1
7
 
Cerf et al. (2010)a [69]; Ferro Cavalcante et al. (2013) [78]; Ferro Cavalcante et al. (2014) [79]; Cerf et al. (2015) [80]
Strakovsky et al. (2011) [81]
de Assis et al. (2006)b [82]
Kjaergaard et al. (2014) [83]
 
Total number of rat studiesa
 
9a
15
17
14
55a
Mice fed high-fat diet for 3 weeks or more prior to pregnancy
Decrease
1
0
7
0
8
 
Mayor et al. (2015) [84]
 
Niculescu and Lupu (2009) [85]; Bentham et al. (2010)b [86]; King et al. (2013) [87]; King et al. (2013)a [88]; Sasson et al. (2015) [89]; Edlow et al. (2016) [90]; Panchenko et al. (2016) [91]
  
No effect
0
2
4
0
6
  
Lager et al. (2014)b [92]; Umekawa et al. (2015) [93]
Liang et al. (2009) [94]; Bytautiene et al. (2011) [95]; King et al. (2013)a [88]; Murabayashi et al. (2013) [96]
  
Increase
2
0
5
0
7
 
Aye et al. (2015) [97]; Rosario et al. (2015) [98]
 
Masuyama and Hiramatsu (2012) [99]; Masuyama and Hiramatsu (2012) [100]; Dahlhoff et al. (2014) [101]; Masuyama and Hiramatsu (2014) [102]; Masuyama et al. (2015) [103]
  
Mice fed high-fat diet for less than 3 weeks prior to pregnancy
Decrease
1
1
1
0
3
 
Sferruzzi-Perri et al. (2013)a [104]
Turdi et al. (2013) [105]
del Mar Plata et al. (2014) [106]
  
No effect
1
2
1
0
4
 
Sferruzzi-Perri et al. (2013) a [104]
Luijten et al. (2013)b [107]; Benatti et al. (2014) [108]
Volpato et al. (2012) [109]
  
Increase
0
1
1
0
2
  
Ashino et al. (2012) [110]
Gregorio et al. (2013) [111]
  
Total number of mouse studiesa
 
4a
6
18a
0
28a
aStudy is listed in the table twice because it included two time points or multiple fat contents with different results. The total number of studies per species counts each study only once
bPercentage of calories from fat was estimated based on energy density of diet, using values from two Research Diets (New Brunswick, NJ) diets (D12451: 45% fat by calories, 4.7 kcal/g, 24% fat by weight, and D12492: 60% fat by calories, 5.21 kcal/g, 35% fat by weight)
The diversity of experimental designs makes comparisons among studies difficult, but studies which include more than one experimental group are useful for examining specific factors that influence the effect on birthweight. One study found that feeding rats diets containing 20 or 30% fat (by energy) from the start of gestation increased birthweight compared with a diet of 10% fat, whereas a 40% fat diet did not [69]. Work by the same group found that a 40% fat diet during the first week of pregnancy reduced birthweight, whereas the same diet fed throughout pregnancy did not [59]. In contrast, other work by this group found that the weight of 20 day-old fetuses was increased by a 40% fat diet administered in third week of gestation, but not by the same diet throughout gestation [80]. Work by a different group found that a cafeteria diet fed to rats for 8 weeks prior to pregnancy reduced fetal weight at day 20, whereas the same diet fed from the start of pregnancy did not [35].
Effects of a maternal HFD may be consistent across gestational ages [89, 93, 101] but may vary through pregnancy. A diet high in fat and simple sugars fed to female mice from the beginning pregnancy reduced fetal weight at G15.5 but not G18.5 [104]. In contrast, a diet with a much higher fat content, and also high in simple sugars, fed to female mice for 12 weeks prior to pregnancy had no effect fetal weight at 14.5 but reduced fetal weight in day 18.5 male fetuses but not female fetuses [88]. Work by the same group found that the same protocol reduced birthweight in females but not males [87]. Inconsistencies in sex-specific effects may arise if the differences between males and females are not tested explicitly, e.g., using a sex by treatment interaction [112]. Studies by a single lab group using the same protocol may yield results that are similar [99, 100, 103] or divergent [66, 77].
Our initial survey did not reveal factors associated with maternal overnutrition that consistently led to increased or decreased fetal growth. We therefore focused more closely on a subset of studies with similar protocols and updated our search. Specifically, we focused on studies using HFDs containing 44–45% fat or 57–62% fat (by energy), as these were relatively numerous. We also restricted this refined search to studies that initiated experimental diets 3 weeks or more prior to pregnancy to include effects of maternal obesity rather than gestational overnutrition alone.
The studies identified in the refined search are summarized in Table 2, and the data extracted from each are provided in Additional file 1: Table S2. Our refined search found only 3 studies that fed mice a 45% fat diet, and all of these reported no effect on birth and/or fetal weight [93, 119, 120]. These studies had very similar protocols, using the same mouse strain (C57BL/6), the same HFD from the same manufacturer, and similar nutrient-matched 10% fat control diets (although these varied in sucrose content). The effect of a 58–60% diet on C57BL/6 mice was more variable, with 7 finding that HFD reduced birth and/or fetal weight [85, 8791, 118], 3 finding no effect [94, 96, 121], and only one finding increased fetal weight [123], although the latter measured fetal weight much earlier than the others (at G12.5). Many of these studies used the same HFD from the same manufacturer, and some used nutrient-controls while others used chow as a control diet. However, even studies using identical HFD and control diets yielded divergent results (e.g., [85, 96] or [90, 123]). Among other mouse strains, the effects of a 60–62% fat diet increased birth and/or fetal weight in 6 studies [99103, 122], although 5 of these were from the same group, and one study found no effect [95]. Among studies feeding 60% fat diets to mice, those finding an increase in fetal growth had fed the experimental diet for a shorter duration prior to pregnancy (4–9 weeks) than those reporting a reduction in fetal growth (9 weeks or more) (Additional file 1: Table S2). There was variability in the sucrose content of the control diets, but this was not associated with effects on birthweight (Additional file 1: Table S2).
Table 2
Summary of studies identified in the updated and refined search
Species
Effect on fetal growth
Calories from fat
Total
~  45%
~  60%
Rat
Decrease
6
1
7
 
Nivoit et al. (2009) [29]; Hayes et al. (2012) [30]; Reynolds et al. (2014) [32]; Dodson et al. (2017) [113]; Huang et al. (2017) [114]; Ye et al. (2017) [115]
Yamada-Obara et al. (2016) [116]
 
No effect
0
7
7
  
Srinivasan et al. (2006) [45]; Gupta et al. (2009) [47]; Mitra et al. (2009) [48]; White et al. (2009) [49]; Guberman et al. (2013) [50]; Desai et al. (2014) [51]; Lecoutre et al. (2016) [117]
 
Increase
1
0
1
 
Song et al. (2015) [55]
  
Total
 
7
8
15
Mouse
Decrease
0
7
7
  
Niculescu and Lupu (2009) [85]; King et al. (2013) [87]; King et al. (2013)a [88]; Sasson et al. (2015) [89]; Edlow et al. (2016) [90]; Panchenko et al. (2016) [91]; Bae-Gartz et al. (2016) [118]
 
No effect
3
5
8
 
Umekawa et al. (2015) [93]; Chin et al. (2017) [119]; Jonscher et al. (2017) [120]
Liang et al. (2009) [94]; Bytautiene et al. (2011) [95]; King et al. (2013)a [88]; Murabayashi et al. (2013) [96]; Connor et al. (2018) [121]
 
Increase
0
7
7
  
Masuyama and Hiramatsu (2012) [99]; Masuyama and Hiramatsu (2012) [100]; Dahlhoff et al. (2014) [101]; Masuyama and Hiramatsu (2014) [102]; Masuyama et al. (2015) [103]; Masuyama et al. (2016) [122]; Nam et al. (2017) [123]
 
Totala
 
3
18a
21a
aStudy is listed in the table twice because it included two time points or multiple fat contents with different results. The total number of studies per species counts each study only once
Rats fed a 45% fat diet generally had offspring with reduced birth and/or fetal weights [29, 30, 32, 113115], although the only study that used a nutrient-matched control diet showed an increase in birthweight [55]. In contrast, feeding rats a 57–60% fat diet generally had no effect on birth or fetal weight [45, 4751, 117], although one study, which also provided fructose in the water of dams on the HFD, found a reduction in birthweight [116]. Variation between studies was not due to strain, as most used Sprague-Dawley rats, and discordant results were observed with this strain.
In both mice and rats fed diets of ~ 45% fat or ~ 60% fat, the HFD generally increased maternal mass prior to conception and impaired glucose tolerance, at least when these parameters were reported (Additional file 1: Table S2). In one case, impaired glucose tolerance was observed in HFD-fed females even though the control diet had a higher sucrose content [96]. Given that mice and rats gestate multiple fetuses per pregnancy, fetal growth might be influenced by effects on litter size, e.g., a reduction in litter size would be expected to increase the growth of individual fetuses. However, most studies that reported litter size found no effects of maternal HFD (Additional file 1: Table S2), and some found reductions in both litter size and the weight of individual offspring [30, 85, 87].

Discussion

Rodents fed HFD are frequently used to study the effects of maternal obesity on offspring health, but we found few consistent effects of maternal HFD on fetal growth. The importance of these findings is not diminished by the fact that rodents are born at a different developmental stage than humans; a rodent model that decreases fetal growth and another that increases fetal growth cannot both model the same human phenotype. Our initial search identified studies with a variety of experimental protocols, and this revealed no clear patterns with respect to how the effects of maternal HFD depend on species, duration of diet, or diet composition. When we refined our focus to studies that fed dams diets containing ~ 45% fat or ~ 60% fat (by energy) for 3 weeks or more prior to pregnancy, we found more consistent results. Mice fed 45% fat diets and rats fed 60% fat diets generally did not show effects on fetal growth. Feeding a 45% fat diet to rats generally reduced birth and fetal weight. However, results were more variable in mice fed a 60% fat diet, with studies feeding the HFD for shorter periods prior to pregnancy (4–9 weeks) more likely to report an increase in fetal growth, whereas those feeding for a longer period were more likely to report a reduction in fetal growth.
Some variability among similar studies is not surprising. Thousands of studies have used rodents fed HFD to study obesity and/or diabetes (not specifically in pregnancy), and effects on weight gain and glycemic control have been inconsistent. Effects depend on diet composition, duration of diet, age and strain of animal, and in some cases vary among different experiments from the same lab [124]. The nature of the control diet is also important, since studies that use chow as controls may differ from a defined HFD in terms of protein source and fiber in addition to fat [124, 125].
A variety of models are needed given that there are a variety of diets and lifestyle characteristics associated with human obesity. However, the translation of the results of animal studies to clinical practice and policy development would benefit from some standardization of models and/or more explicit relation of the model to a particular human phenotype. Below, we discuss three key issues to consider in the design of experiments to examine the long-term consequences of maternal overnutrition and/ or obesity, and provide recommendations for standardized models.

Experimental diets and controls

The percentage of energy from fat in US diets is estimated to be in the range of 30–40% in obese and normal weight individuals [124, 126, 127], and therefore experimental diets with much more than 40% of energy from fat may provide weaker models of human pathophysiology. Similarly, the median percentage of calories from protein in US diets is 14% [128], whereas many of the studies that we identified used purified HFDs that were 20% protein, although some attempted to mimic the protein content of Western diets [113, 129]. Thus, while many experimental diets are successful in increasing maternal pre-pregnancy weight and/ or impairing glucose metabolism, the resulting obesity may not accurately reflect common human phenotypes. To facilitate putting experimental diets into a human context, studies should be required to report the macronutrient composition of experimental and control diets as a percentage of total energy, rather than only reporting contents by weight and/ or the overall energy density.
Although we focused on studies that provided a HFD ad libitum for a specific period prior to and/or during pregnancy, we acknowledge that other experimental approaches offer advantages for studying the effects of maternal obesity. Embryo transfers have been used to distinguish the effects of pre-gestational and gestational exposure to maternal obesity [89]. Shankar et al. [130] fed liquid diets to rats by intragastric cannulation to induce obesity by controlling the number of calories provided. A contrasting approach is to control food intake to keep energy consumption similar and vary only the macronutrient composition of the maternal diet [131]. To ensure that the HFD induces maternal obesity, some studies mate HFD females when their body weight has increased by a certain amount, and compare these with age-matched females on a control diet. In mice, this approach increased fetal weight in two studies [97, 98], had no effect in another [132], and decreased fetal weight in a fourth [90]. Ye et al. [115] fed rats a HFD and selected those with the greatest weight gain (susceptible to diet-induced obesity) and those with the lowest weight gain (resistant to diet-induced obesity) and found that fetal weight was reduced in susceptible dams but not in resistant dams.

Effects on birthweight

It is concerning that a substantial number of studies found that a maternal HFD reduced fetal growth, because this is not a typical feature of obese pregnancy in humans. Although some reviews suggest that obese mothers are at increased risk of delivering small infants, closer scrutiny of the primary literature cited does not support such claims. To the contrary, some studies have found that obesity reduces the risk of delivering a small-for-gestational age or low-birthweight baby [133136]. Obesity increases the risk of preeclampsia, and preeclampsia is often associated with intrauterine growth restriction [11, 137] but it does not necessarily follow that obesity increases the risk of intrauterine growth restriction. Studies reporting increased risk of preeclampsia in obese pregnancies generally do not find associations between obesity and IUGR [138, 139]. We know of only one study that found an increased risk of having a small-for-gestational-age infant among obese women, and this was found only in association with a body mass index (BMI) over 40, and not with a BMI of 29–40 [140]. The association was not significant after removing women with preeclampsia [140], suggesting that obesity specifically increased the risk of growth restriction in preeclamptic women. Thus, apart from this study, there is little evidence that human obesity is associated with fetal growth restriction. Therefore, experimental approaches that result in reduced fetal growth are less relevant as models of obesity and overnutrition in humans, and it may be useful to avoid the protocols that we have identified as consistently reducing fetal growth. Based on developmental milestones, rodent birthweight only provides a model of human fetal growth up to the end of the second trimester. However, it is unlikely that a rodent model with reduced birthweight is a good model for an obese human pregnancy with an increased risk of high birthweight, unless it can be shown that human obesity results in growth restriction early in pregnancy that recovers in the last trimester. Experimental approaches that increase fetal growth are suitable for modeling high birthweights, which occurs at higher frequency in obese pregnancies [6, 10]. Mating HFD mice when their weight had increased by 25% led to higher fetal weight in two studies [97, 98] but had no effect in another [132]. While the solid component of the HFD was 41% fat in these studies, the provision of a sucrose solution with the HFD would have reduced the percentage of calories from fat (and protein), perhaps to levels more typical of Western diets. Protocols with no effect on fetal growth may be useful for studying effects of maternal obesity on offspring health that occur independently of birthweight [141].

Species and strain

As model species, rats and mice offer different strengths and weaknesses. The larger size of rats facilitates various physiological manipulations and measurements, while mice are more amenable to genetic manipulation, and their smaller size reduces cost per animal [142]. Rat strains are generally outbred (Wistar, Sprague Dawley, Long-Evans) while the most commonly used mouse strains (e.g., C57BL/6) are inbred [143]. While inbred strains reduce variability and thereby increase the power to detect certain effects, outbred strains may provide a better model of the genetic heterogeneity of human populations [25], and may also be of greater relevance for effects dependent on the immune system [142].
In a recent meta-analysis, maternal HFD tended to increase birthweight in mice and decrease birthweight in rats, although species and strain contributed relatively little to between-study variability in the metabolic effects of maternal HFD [27]. Strains of mice differ in their susceptibility to the metabolic effects of HFD [144], but it is not necessarily the case that more susceptible strains are better models; more resistant strains may offer the opportunity to study differences between obesity-susceptible and obesity-resistant individuals (e.g., [115]). Glucose metabolism even differs between substrains of C57BL/6 [145], which underscores the importance of reporting the substrain, e.g., C57BL/6 J vs. C57BL/6 N.
While mice and rats are frequently used to investigate the effects of maternal nutrition, these species are born at a substantially different developmental stage than humans and generally have multiple fetuses per pregnancy. In these and some other respects, other rodent models such as guinea pigs offer advantages over mice and rats [146]. A more complete consideration of species selection is provided elsewhere [142, 147]. As with the diet protocol, the rationale for the choice of species and the specific situation it attempts to model should be described explicitly [142]. Nevertheless, there are broad similarities in the phenotypes of offspring exposed to maternal HFD between mice, rats and non-human primates [143].

Conclusions

The effects of HFD during pregnancy have been examined with a diverse array of experimental protocols yielding few clear patterns with respect to approaches that increase or decrease fetal growth. However, studies with similar protocols using the same strain of animal yield more consistent results. Even so, the HFD used most frequently with rodents do not closely match Western diets, the former being higher in fat and protein. Perhaps for this reason, many studies find that a maternal HFD reduces fetal growth in rodents, which is not a typical feature of obese human pregnancies. The present review has identified experimental approaches that increase birthweight or have no effect, and the adoption and standardization of such protocols will improve the translational impact of research into the effects of maternal overnutrition on offspring health.

Acknowledgements

None.

Funding

This study was funded by a Natural Sciences and Engineering Research Council of Canada Discovery Grant (JKC; grant number RGPIN-2016-04047) and a Simon Fraser University Vice President, Research Undergraduate Student Research Award (KIL). The funding bodies had no role in study design, collection and interpretation of data or in manuscript preparation.

Availability of data and materials

This review was based on published data.
This review study was based on published work and therefore did not require approval by an institutional committee.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Hoffman DJ, Reynolds RM, Hardy DB. Developmental origins of health and disease: current knowledge and potential mechanisms. Nutr Rev. 2017;75:951–70.CrossRefPubMed Hoffman DJ, Reynolds RM, Hardy DB. Developmental origins of health and disease: current knowledge and potential mechanisms. Nutr Rev. 2017;75:951–70.CrossRefPubMed
2.
Zurück zum Zitat Barker DJP, Osmond C, Kajantie E, Eriksson JG. Growth and chronic disease: findings in the Helsinki birth cohort. Ann Hum Biol. 2009;36:444–58.CrossRef Barker DJP, Osmond C, Kajantie E, Eriksson JG. Growth and chronic disease: findings in the Helsinki birth cohort. Ann Hum Biol. 2009;36:444–58.CrossRef
3.
Zurück zum Zitat Barker DJP, Osmond C, Winter PD, Margetts B, Simmonds SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989;334:577–80.CrossRef Barker DJP, Osmond C, Winter PD, Margetts B, Simmonds SJ. Weight in infancy and death from ischaemic heart disease. Lancet. 1989;334:577–80.CrossRef
5.
Zurück zum Zitat Hales CN, Barker DJP, Clark PMS, Cox LJ, Osmond C, Winter PD. Fetal and infant growth and impaired glucose tolerance at age 64. Br Med J. 1991;303:1019–22.CrossRef Hales CN, Barker DJP, Clark PMS, Cox LJ, Osmond C, Winter PD. Fetal and infant growth and impaired glucose tolerance at age 64. Br Med J. 1991;303:1019–22.CrossRef
6.
Zurück zum Zitat Sen S, Carpenter AH, Hochstadt J, Huddleston JY, Kustanovich V, Reynolds AA, et al. Nutrition, weight gain and eating behavior in pregnancy: a review of experimental evidence for long-term effects on the risk of obesity in offspring. Physiol Behav. 2012;107:138–45.CrossRefPubMed Sen S, Carpenter AH, Hochstadt J, Huddleston JY, Kustanovich V, Reynolds AA, et al. Nutrition, weight gain and eating behavior in pregnancy: a review of experimental evidence for long-term effects on the risk of obesity in offspring. Physiol Behav. 2012;107:138–45.CrossRefPubMed
7.
Zurück zum Zitat Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiol Rev. 2014;94:1027–76.CrossRefPubMedPubMedCentral Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiol Rev. 2014;94:1027–76.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Grissom NM, Reyes TM. Gestational overgrowth and undergrowth affect neurodevelopment: similarities and differences from behavior to epigenetics. Int J Dev Neurosci. 2013;31:406–14.CrossRefPubMed Grissom NM, Reyes TM. Gestational overgrowth and undergrowth affect neurodevelopment: similarities and differences from behavior to epigenetics. Int J Dev Neurosci. 2013;31:406–14.CrossRefPubMed
9.
Zurück zum Zitat Li M, Sloboda DM, Vickers MH. Maternal obesity and developmental programming of metabolic disorders in offspring: evidence from animal models. Exp Diabetes Res. 2011;2011:592408.CrossRefPubMedPubMedCentral Li M, Sloboda DM, Vickers MH. Maternal obesity and developmental programming of metabolic disorders in offspring: evidence from animal models. Exp Diabetes Res. 2011;2011:592408.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Yu Z, Han S, Zhu J, Sun X, Ji C, Guo X. 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:e61627.CrossRefPubMedPubMedCentral Yu Z, Han S, Zhu J, Sun X, Ji C, Guo X. 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:e61627.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Howell KR, Powell TL. Effects of maternal obesity on placental function and fetal development. Reproduction. 2017;153:R97–108.CrossRefPubMed Howell KR, Powell TL. Effects of maternal obesity on placental function and fetal development. Reproduction. 2017;153:R97–108.CrossRefPubMed
12.
Zurück zum Zitat Dennedy MC, Avalos G, O’Reilly MW, O’Sullivan EP, Gaffney G, Dunne F. ATLANTIC-DIP: raised maternal body mass index (BMI) adversely affects maternal and fetal outcomes in glucose-tolerant women according to International Association of Diabetes and Pregnancy Study Groups (IADPSG) criteria. J Clin Endocrinol Metab. 2012;97:E608–12.CrossRefPubMed Dennedy MC, Avalos G, O’Reilly MW, O’Sullivan EP, Gaffney G, Dunne F. ATLANTIC-DIP: raised maternal body mass index (BMI) adversely affects maternal and fetal outcomes in glucose-tolerant women according to International Association of Diabetes and Pregnancy Study Groups (IADPSG) criteria. J Clin Endocrinol Metab. 2012;97:E608–12.CrossRefPubMed
13.
Zurück zum Zitat Grieger JA, Clifton VL. A review of the impact of dietary intakes in human pregnancy on infant birthweight. Nutrients. 2015;7:153–78.CrossRef Grieger JA, Clifton VL. A review of the impact of dietary intakes in human pregnancy on infant birthweight. Nutrients. 2015;7:153–78.CrossRef
14.
Zurück zum Zitat Stang J, Huffman LG. Position of the academy of nutrition and dietetics: obesity, reproduction, and pregnancy outcomes. J Acad Nutr Diet. 2016;116:677–91.CrossRefPubMed Stang J, Huffman LG. Position of the academy of nutrition and dietetics: obesity, reproduction, and pregnancy outcomes. J Acad Nutr Diet. 2016;116:677–91.CrossRefPubMed
15.
Zurück zum Zitat Tanentsapf I, Heitmann BL, Adegboye ARA. Systematic review of clinical trials on dietary interventions to prevent excessive weight gain during pregnancy among normal weight, overweight and obese women. BMC Pregnancy Childbirth. 2011;11:81.CrossRefPubMedPubMedCentral Tanentsapf I, Heitmann BL, Adegboye ARA. Systematic review of clinical trials on dietary interventions to prevent excessive weight gain during pregnancy among normal weight, overweight and obese women. BMC Pregnancy Childbirth. 2011;11:81.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Nathanielsz PW, Ford SP, Long NM, Vega CC, Reyes-Castro LA, Zambrano E. Interventions to prevent adverse fetal programming due to maternal obesity during pregnancy. Nutr Rev. 2013;71:S78–87.CrossRefPubMed Nathanielsz PW, Ford SP, Long NM, Vega CC, Reyes-Castro LA, Zambrano E. Interventions to prevent adverse fetal programming due to maternal obesity during pregnancy. Nutr Rev. 2013;71:S78–87.CrossRefPubMed
17.
Zurück zum Zitat 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;10:47.CrossRefPubMedPubMedCentral 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;10:47.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Gresham E, Byles JE, Bisquera A, Hure AJ. Effects of dietary interventions on neonatal and infant outcomes: a systematic review and meta-analysis. Am J Clin Nutr. 2014;100:1298–321.CrossRefPubMed Gresham E, Byles JE, Bisquera A, Hure AJ. Effects of dietary interventions on neonatal and infant outcomes: a systematic review and meta-analysis. Am J Clin Nutr. 2014;100:1298–321.CrossRefPubMed
19.
Zurück zum Zitat Murphy MM, Stettler N, Smith KM, Reiss R. Associations of consumption of fruits and vegetables during pregnancy with infant birth weight or small for gestational age births: a systematic review of the literature. Int J Women's Health. 2014;6:899–912.CrossRef Murphy MM, Stettler N, Smith KM, Reiss R. Associations of consumption of fruits and vegetables during pregnancy with infant birth weight or small for gestational age births: a systematic review of the literature. Int J Women's Health. 2014;6:899–912.CrossRef
20.
Zurück zum Zitat McGee M, Bainbridge S, Fontaine-Bisson B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev. 2018;76:469–78.CrossRefPubMed McGee M, Bainbridge S, Fontaine-Bisson B. A crucial role for maternal dietary methyl donor intake in epigenetic programming and fetal growth outcomes. Nutr Rev. 2018;76:469–78.CrossRefPubMed
21.
Zurück zum Zitat Pannia E, Cho CE, Kubant R, Sanchez-Hernandez D, Huot PSP, Anderson GH. Role of maternal vitamins in programming health and chronic disease. Nutr Rev. 2016;74:166–80.CrossRefPubMedPubMedCentral Pannia E, Cho CE, Kubant R, Sanchez-Hernandez D, Huot PSP, Anderson GH. Role of maternal vitamins in programming health and chronic disease. Nutr Rev. 2016;74:166–80.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Lewis RM, Demmelmair H, Gaillard R, Godfrey KM, Hauguel-De Mouzon S, Huppertz B, et al. The placental exposome: Placental determinants of fetal adiposity and postnatal body composition. Ann Nutr Metab. 2013;63:208–15. Lewis RM, Demmelmair H, Gaillard R, Godfrey KM, Hauguel-De Mouzon S, Huppertz B, et al. The placental exposome: Placental determinants of fetal adiposity and postnatal body composition. Ann Nutr Metab. 2013;63:208–15.
23.
Zurück zum Zitat Ainge H, Thompson C, Ozanne SE, Rooney KB. A systematic review on animal models of maternal high fat feeding and offspring glycaemic control. Int J Obes. 2011;35:325–35.CrossRef Ainge H, Thompson C, Ozanne SE, Rooney KB. A systematic review on animal models of maternal high fat feeding and offspring glycaemic control. Int J Obes. 2011;35:325–35.CrossRef
24.
Zurück zum Zitat Blackmore HL, Ozanne SE. Maternal diet-induced obesity and offspring cardiovascular health. J Dev Orig Health Dis. 2013;4:338–47.CrossRefPubMed Blackmore HL, Ozanne SE. Maternal diet-induced obesity and offspring cardiovascular health. J Dev Orig Health Dis. 2013;4:338–47.CrossRefPubMed
25.
Zurück zum Zitat Morton JS, Cooke C-L, Davidge ST. In utero origins of hypertension: mechanisms and targets for therapy. Physiol Rev. 2016;96:549–603.CrossRefPubMed Morton JS, Cooke C-L, Davidge ST. In utero origins of hypertension: mechanisms and targets for therapy. Physiol Rev. 2016;96:549–603.CrossRefPubMed
26.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
27.
Zurück zum Zitat Ribaroff GA, Wastnedge E, Drake AJ, Sharpe RM, Chambers TJG. Animal models of maternal high fat diet exposure and effects on metabolism in offspring: a meta-regression analysis. Obes Rev. 2017;18:673–86. Ribaroff GA, Wastnedge E, Drake AJ, Sharpe RM, Chambers TJG. Animal models of maternal high fat diet exposure and effects on metabolism in offspring: a meta-regression analysis. Obes Rev. 2017;18:673–86.
28.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009;6:e1000097.CrossRefPubMedPubMedCentral Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009;6:e1000097.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Nivoit P, Morens C, Van Assche FA, Jansen E, Poston L, Remacle C, et al. Established diet-induced obesity in female rats leads to offspring hyperphagia, adiposity and insulin resistance. Diabetologia. 2009;52:1133–42.CrossRefPubMed Nivoit P, Morens C, Van Assche FA, Jansen E, Poston L, Remacle C, et al. Established diet-induced obesity in female rats leads to offspring hyperphagia, adiposity and insulin resistance. Diabetologia. 2009;52:1133–42.CrossRefPubMed
30.
Zurück zum Zitat Hayes EK, Lechowicz A, Petrik JJ, Storozhuk Y, Paez-Parent S, Dai Q, et al. Adverse fetal and neonatal outcomes associated with a life-long high fat diet: role of altered development of the placental vasculature. PLoS One. 2012;7:e33370.CrossRefPubMedPubMedCentral Hayes EK, Lechowicz A, Petrik JJ, Storozhuk Y, Paez-Parent S, Dai Q, et al. Adverse fetal and neonatal outcomes associated with a life-long high fat diet: role of altered development of the placental vasculature. PLoS One. 2012;7:e33370.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Christante CM, Taboga SR, Pinto-Fochi ME, Goes RM. Maternal obesity disturbs the postnatal development of gonocytes in the rat without impairment of testis structure at prepubertal age. Reproduction. 2013;146:549–58.CrossRefPubMed Christante CM, Taboga SR, Pinto-Fochi ME, Goes RM. Maternal obesity disturbs the postnatal development of gonocytes in the rat without impairment of testis structure at prepubertal age. Reproduction. 2013;146:549–58.CrossRefPubMed
32.
Zurück zum Zitat Reynolds CM, Vickers MH, Harrison CJ, Segovia SA, Gray C. High fat and/or high salt intake during pregnancy alters maternal meta-inflammation and offspring growth and metabolic profiles. Physiol Rep. 2014;2:e12110.CrossRefPubMedPubMedCentral Reynolds CM, Vickers MH, Harrison CJ, Segovia SA, Gray C. High fat and/or high salt intake during pregnancy alters maternal meta-inflammation and offspring growth and metabolic profiles. Physiol Rep. 2014;2:e12110.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Couvreur O, Ferezou J, Gripois D, Serougne C, Crepin D, Aubourg A, et al. Unexpected Long-term protection of adult offspring born to high-fat fed dams against obesity induced by a sucrose-rich diet. PLoS One. 2011;6:e18043.CrossRefPubMedPubMedCentral Couvreur O, Ferezou J, Gripois D, Serougne C, Crepin D, Aubourg A, et al. Unexpected Long-term protection of adult offspring born to high-fat fed dams against obesity induced by a sucrose-rich diet. PLoS One. 2011;6:e18043.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Li S, Tse IMY, Li ETS. Maternal green tea extract supplementation to rats fed a high-fat diet ameliorates insulin resistance in adult male offspring. J Nutr Biochem. 2012;23:1655–60.CrossRefPubMed Li S, Tse IMY, Li ETS. Maternal green tea extract supplementation to rats fed a high-fat diet ameliorates insulin resistance in adult male offspring. J Nutr Biochem. 2012;23:1655–60.CrossRefPubMed
35.
Zurück zum Zitat Akyol A, Langley-Evans SC, McMullen S. Obesity induced by cafeteria feeding and pregnancy outcome in the rat. Br J Nutr. 2009;102:1601–10.CrossRefPubMed Akyol A, Langley-Evans SC, McMullen S. Obesity induced by cafeteria feeding and pregnancy outcome in the rat. Br J Nutr. 2009;102:1601–10.CrossRefPubMed
36.
Zurück zum Zitat Raipuria M, Bahari H, Morris MJ. Effects of maternal diet and exercise during pregnancy on glucose metabolism in skeletal muscle and fat of weanling rats. PLoS One. 2015;10:e0120980.CrossRefPubMedPubMedCentral Raipuria M, Bahari H, Morris MJ. Effects of maternal diet and exercise during pregnancy on glucose metabolism in skeletal muscle and fat of weanling rats. PLoS One. 2015;10:e0120980.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Ramirez-Lopez MT, Vazquez M, Bindila L, Lomazzo E, Hofmann C, Blanco RN, et al. Exposure to a highly caloric palatable diet during Pregestational and gestational periods affects hypothalamic and hippocampal endocannabinoid levels at birth and induces adiposity and anxiety-like behaviors in male rat offspring. Front Behav Neurosci. 2016;9:339.CrossRefPubMedPubMedCentral Ramirez-Lopez MT, Vazquez M, Bindila L, Lomazzo E, Hofmann C, Blanco RN, et al. Exposure to a highly caloric palatable diet during Pregestational and gestational periods affects hypothalamic and hippocampal endocannabinoid levels at birth and induces adiposity and anxiety-like behaviors in male rat offspring. Front Behav Neurosci. 2016;9:339.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Sánchez-Blanco C, Amusquivar E, Bispo K, Herrera E. Influence of cafeteria diet and fish oil in pregnancy and lactation on pups’ body weight and fatty acid profiles in rats. Eur J Nutr. 2016;55:1741–53.CrossRefPubMed Sánchez-Blanco C, Amusquivar E, Bispo K, Herrera E. Influence of cafeteria diet and fish oil in pregnancy and lactation on pups’ body weight and fatty acid profiles in rats. Eur J Nutr. 2016;55:1741–53.CrossRefPubMed
39.
Zurück zum Zitat DelPrado 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;127:458–62.CrossRef DelPrado 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;127:458–62.CrossRef
40.
Zurück zum Zitat Caluwaerts S, Lambin S, van Bree R, Peeters H, Vergote I, Verhaeghe J. Diet-induced obesity in gravid rats engenders early hyperadiposity in the offspring. Metab Exp. 2007;56:1431–8.CrossRef Caluwaerts S, Lambin S, van Bree R, Peeters H, Vergote I, Verhaeghe J. Diet-induced obesity in gravid rats engenders early hyperadiposity in the offspring. Metab Exp. 2007;56:1431–8.CrossRef
41.
Zurück zum Zitat Nitert MD, Vaswani K, Hum M, Chan H-W, Wood-Bradley R, Henry S, et al. Maternal high-fat diet alters expression of pathways of growth, blood supply and arachidonic acid in rat placenta. J Nutr Sci. 2013;2:e41.CrossRef Nitert MD, Vaswani K, Hum M, Chan H-W, Wood-Bradley R, Henry S, et al. Maternal high-fat diet alters expression of pathways of growth, blood supply and arachidonic acid in rat placenta. J Nutr Sci. 2013;2:e41.CrossRef
42.
Zurück zum Zitat Zambrano E, Martinez-Samayoa PM, Rodriguez-Gonzalez GL, Nathanielsz PW. Dietary intervention prior to pregnancy reverses metabolic programming in male offspring of obese rats. J Physiol. 2010;588:1791–9.CrossRefPubMedPubMedCentral Zambrano E, Martinez-Samayoa PM, Rodriguez-Gonzalez GL, Nathanielsz PW. Dietary intervention prior to pregnancy reverses metabolic programming in male offspring of obese rats. J Physiol. 2010;588:1791–9.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Vega CC, Reyes-Castro LA, Bautista CJ, Larrea F, Nathanielsz PW, Zambrano E. Exercise in obese female rats has beneficial effects on maternal and male and female offspring metabolism. Int J Obes. 2015;39:712–9.CrossRef Vega CC, Reyes-Castro LA, Bautista CJ, Larrea F, Nathanielsz PW, Zambrano E. Exercise in obese female rats has beneficial effects on maternal and male and female offspring metabolism. Int J Obes. 2015;39:712–9.CrossRef
44.
Zurück zum Zitat Shaw MA, Rasmussen KM, Myers TR. Consumption of a high fat diet impairs reproductive performance in Sprague-Dawley rats. J Nutr. 1997;127:64–9.CrossRefPubMed Shaw MA, Rasmussen KM, Myers TR. Consumption of a high fat diet impairs reproductive performance in Sprague-Dawley rats. J Nutr. 1997;127:64–9.CrossRefPubMed
45.
Zurück zum Zitat Srinivasan M, Katewa SD, Palaniyappan A, Pandya JD, Patel MS. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Metab. 2006;291:E792–9. Srinivasan M, Katewa SD, Palaniyappan A, Pandya JD, Patel MS. Maternal high-fat diet consumption results in fetal malprogramming predisposing to the onset of metabolic syndrome-like phenotype in adulthood. Am J Physiol Metab. 2006;291:E792–9.
46.
Zurück zum Zitat Ferezou-Viala J, Roy A-F, Serougne C, Gripois D, Parquet M, Bailleux V, et al. Long-term consequences of maternal high-fat feeding on hypothalamic leptin sensitivity and diet-induced obesity in the offspring. Am J Physiol Integr Comp Physiol. 2007;293:R1056–62.CrossRef Ferezou-Viala J, Roy A-F, Serougne C, Gripois D, Parquet M, Bailleux V, et al. Long-term consequences of maternal high-fat feeding on hypothalamic leptin sensitivity and diet-induced obesity in the offspring. Am J Physiol Integr Comp Physiol. 2007;293:R1056–62.CrossRef
47.
Zurück zum Zitat Gupta A, Srinivasan M, Thamadilok S, Patel MS. Hypothalamic alterations in fetuses of high fat diet-fed obese female rats. J Endocrinol. 2009;200:293–300.CrossRefPubMed Gupta A, Srinivasan M, Thamadilok S, Patel MS. Hypothalamic alterations in fetuses of high fat diet-fed obese female rats. J Endocrinol. 2009;200:293–300.CrossRefPubMed
48.
Zurück zum Zitat Mitra A, Alvers KM, Crump EM, Rowland NE. Effect of high-fat diet during gestation, lactation, or postweaning on physiological and behavioral indexes in borderline hypertensive rats. Am J Physiol Integr Comp Physiol. 2009;296:R20–8.CrossRef Mitra A, Alvers KM, Crump EM, Rowland NE. Effect of high-fat diet during gestation, lactation, or postweaning on physiological and behavioral indexes in borderline hypertensive rats. Am J Physiol Integr Comp Physiol. 2009;296:R20–8.CrossRef
49.
Zurück zum Zitat White CL, Purpera MN, Morrison CD. Maternal obesity is necessary for programming effect of high-fat diet on offspring. Am J Physiol Integr Comp Physiol. 2009;296:R1464–72.CrossRef White CL, Purpera MN, Morrison CD. Maternal obesity is necessary for programming effect of high-fat diet on offspring. Am J Physiol Integr Comp Physiol. 2009;296:R1464–72.CrossRef
50.
Zurück zum Zitat Guberman C, Jellyman JK, Han G, Ross MG, Desai M. Maternal high-fat diet programs rat offspring hypertension and activates the adipose renin-angiotensin system. Am J Obstet Gynecol. 2013;209:262.e1–8.CrossRef Guberman C, Jellyman JK, Han G, Ross MG, Desai M. Maternal high-fat diet programs rat offspring hypertension and activates the adipose renin-angiotensin system. Am J Obstet Gynecol. 2013;209:262.e1–8.CrossRef
51.
Zurück zum Zitat Desai M, Jellyman JK, Han G, Beall M, Lane RH, Ross MG. Maternal obesity and high-fat diet program offspring metabolic syndrome. Am J Obstet Gynecol. 2014;211:237.e1–237.e13.CrossRef Desai M, Jellyman JK, Han G, Beall M, Lane RH, Ross MG. Maternal obesity and high-fat diet program offspring metabolic syndrome. Am J Obstet Gynecol. 2014;211:237.e1–237.e13.CrossRef
52.
Zurück zum Zitat Rolls BJ, Rowe EA. Pregnancy and lactation in the obese rat - effects on maternal and pup weights. Physiol Behav. 1982;28:393–400.CrossRefPubMed Rolls BJ, Rowe EA. Pregnancy and lactation in the obese rat - effects on maternal and pup weights. Physiol Behav. 1982;28:393–400.CrossRefPubMed
53.
Zurück zum Zitat Chen H, Simar D, Lambert K, Mercier J, Morris MJ. Maternal and postnatal Overnutrition differentially impact appetite regulators and fuel metabolism. Endocrinology. 2008;149:5348–56.CrossRefPubMed Chen H, Simar D, Lambert K, Mercier J, Morris MJ. Maternal and postnatal Overnutrition differentially impact appetite regulators and fuel metabolism. Endocrinology. 2008;149:5348–56.CrossRefPubMed
54.
Zurück zum Zitat 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;25:2167–79.CrossRefPubMedPubMedCentral 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;25:2167–79.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Song Y, Yu Y, Wang D, Chai S, Liu D, Xiao X, et al. Maternal high-fat diet feeding during pregnancy and lactation augments lung inflammation and remodeling in the offspring. Respir Physiol Neurobiol. 2015;207:1–6.CrossRefPubMed Song Y, Yu Y, Wang D, Chai S, Liu D, Xiao X, et al. Maternal high-fat diet feeding during pregnancy and lactation augments lung inflammation and remodeling in the offspring. Respir Physiol Neurobiol. 2015;207:1–6.CrossRefPubMed
56.
Zurück zum Zitat Gaccioli F, White V, Capobianco E, Powell TL, Jawerbaum A, Jansson T. Maternal overweight induced by a diet with high content of saturated fat activates placental mTOR and eIF2alpha signaling and increases fetal growth in rats. Biol Reprod. 2013;89:96.CrossRefPubMed Gaccioli F, White V, Capobianco E, Powell TL, Jawerbaum A, Jansson T. Maternal overweight induced by a diet with high content of saturated fat activates placental mTOR and eIF2alpha signaling and increases fetal growth in rats. Biol Reprod. 2013;89:96.CrossRefPubMed
57.
Zurück zum Zitat Mazzucco MB, Higa R, Capobianco E, Kurtz M, Jawerbaum A, White V. Saturated fat-rich diet increases fetal lipids and modulates LPL and leptin receptor expression in rat placentas. J Endocrinol. 2013;217:303–15.CrossRefPubMed Mazzucco MB, Higa R, Capobianco E, Kurtz M, Jawerbaum A, White V. Saturated fat-rich diet increases fetal lipids and modulates LPL and leptin receptor expression in rat placentas. J Endocrinol. 2013;217:303–15.CrossRefPubMed
58.
Zurück zum Zitat Yang X, Lai P, Chen X-G, Liu Z-P, Tang Y-M, Rao S, et al. Maternal high-fat diet exposure leads to insulin resistance and impacts myogenic and adipogenicgene expression in offspring rats. Eur J Lipid Sci Technol. 2015;117:1550–60.CrossRef Yang X, Lai P, Chen X-G, Liu Z-P, Tang Y-M, Rao S, et al. Maternal high-fat diet exposure leads to insulin resistance and impacts myogenic and adipogenicgene expression in offspring rats. Eur J Lipid Sci Technol. 2015;117:1550–60.CrossRef
59.
Zurück zum Zitat Cerf ME, Williams K, Nkomo XI, Muller CJ, Du Toit DF, Louw J, et al. Islet cell response in the neonatal rat after exposure to a high-fat diet during pregnancy. Am J Physiol Integr Comp Physiol. 2005;288:R1122–8.CrossRef Cerf ME, Williams K, Nkomo XI, Muller CJ, Du Toit DF, Louw J, et al. Islet cell response in the neonatal rat after exposure to a high-fat diet during pregnancy. Am J Physiol Integr Comp Physiol. 2005;288:R1122–8.CrossRef
60.
Zurück zum Zitat Mark PJ, Sisala C, Connor K, Patel R, Lewis JL, Vickers MH, et al. A maternal high-fat diet in rat pregnancy reduces growth of the fetus and the placental junctional zone, but not placental labyrinth zone growth. J Dev Orig Health Dis. 2011;2:63–70.CrossRef Mark PJ, Sisala C, Connor K, Patel R, Lewis JL, Vickers MH, et al. A maternal high-fat diet in rat pregnancy reduces growth of the fetus and the placental junctional zone, but not placental labyrinth zone growth. J Dev Orig Health Dis. 2011;2:63–70.CrossRef
61.
Zurück zum Zitat Smith T, Sloboda DM, Saffery R, Joo E, Vickers MH. Maternal nutritional history modulates the hepatic IGF-IGFBP axis in adult male rat offspring. Endocrine. 2014;46:70–82.CrossRefPubMed Smith T, Sloboda DM, Saffery R, Joo E, Vickers MH. Maternal nutritional history modulates the hepatic IGF-IGFBP axis in adult male rat offspring. Endocrine. 2014;46:70–82.CrossRefPubMed
62.
Zurück zum Zitat Cordero P, Gonzalez-Muniesa P, Milagro FI, Campion J, Martinez JA. Perinatal maternal feeding with an energy dense diet and/or micronutrient mixture drives offspring fat distribution depending on the sex and growth stage. J Anim Physiol Anim Nutr (Berl). 2015;99:834–40.CrossRef Cordero P, Gonzalez-Muniesa P, Milagro FI, Campion J, Martinez JA. Perinatal maternal feeding with an energy dense diet and/or micronutrient mixture drives offspring fat distribution depending on the sex and growth stage. J Anim Physiol Anim Nutr (Berl). 2015;99:834–40.CrossRef
63.
Zurück zum Zitat da Silva Cunha F, Molle RD, Portella AK, da Silva BC, Noschang C, Goldani MZ, et al. Both food restriction and high-fat diet during gestation induce low birth weight and altered physical activity in adult rat offspring: the “similarities in the inequalities” model. PLoS One. 2015;10:e0118586. da Silva Cunha F, Molle RD, Portella AK, da Silva BC, Noschang C, Goldani MZ, et al. Both food restriction and high-fat diet during gestation induce low birth weight and altered physical activity in adult rat offspring: the “similarities in the inequalities” model. PLoS One. 2015;10:e0118586.
64.
Zurück zum Zitat Segovia SA, Vickers MH, Zhang XD, Gray C, Reynolds CM. Maternal supplementation with conjugated linoleic acid in the setting of diet-induced obesity normalises the inflammatory phenotype in mothers and reverses metabolic dysfunction and impaired insulin sensitivity in offspring. J Nutr Biochem. 2015;26:1448–57.CrossRefPubMed Segovia SA, Vickers MH, Zhang XD, Gray C, Reynolds CM. Maternal supplementation with conjugated linoleic acid in the setting of diet-induced obesity normalises the inflammatory phenotype in mothers and reverses metabolic dysfunction and impaired insulin sensitivity in offspring. J Nutr Biochem. 2015;26:1448–57.CrossRefPubMed
65.
Zurück zum Zitat 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;98:843–51.CrossRefPubMed 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;98:843–51.CrossRefPubMed
66.
Zurück zum Zitat Zhang Z-Y, Zeng J-J, Kjaergaard M, Guan N, Raun K, Nilsson C, et al. Effects of a maternal diet supplemented with chocolate and fructose beverage during gestation and lactation on rat dams and their offspring. Clin Exp Pharmacol Physiol. 2011;38:613–22.CrossRefPubMed Zhang Z-Y, Zeng J-J, Kjaergaard M, Guan N, Raun K, Nilsson C, et al. Effects of a maternal diet supplemented with chocolate and fructose beverage during gestation and lactation on rat dams and their offspring. Clin Exp Pharmacol Physiol. 2011;38:613–22.CrossRefPubMed
67.
Zurück zum Zitat Gugusheff JR, Ong ZY, Muhlhausler BS. A maternal “junk-food” diet reduces sensitivity to the opioid antagonist naloxone in offspring postweaning. FASEB J. 2013;27:1275–84.CrossRefPubMed Gugusheff JR, Ong ZY, Muhlhausler BS. A maternal “junk-food” diet reduces sensitivity to the opioid antagonist naloxone in offspring postweaning. FASEB J. 2013;27:1275–84.CrossRefPubMed
68.
Zurück zum Zitat Martin JC, Martin DC, Shores P, Chao S. Growth and activity but not maturation are affected by perinatal diet. Physiol Behav. 1987;40:279–85.CrossRefPubMed Martin JC, Martin DC, Shores P, Chao S. Growth and activity but not maturation are affected by perinatal diet. Physiol Behav. 1987;40:279–85.CrossRefPubMed
69.
Zurück zum Zitat Cerf ME, Williams K, van Rooyen J, Esterhuyse AJ, Muller CJ, Louw J. Gestational 30 and 40% fat diets increase brain GLUT2 and neuropeptide Y immunoreactivity in neonatal Wistar rats. Int J Dev Neurosci. 2010;28:625–30.CrossRefPubMed Cerf ME, Williams K, van Rooyen J, Esterhuyse AJ, Muller CJ, Louw J. Gestational 30 and 40% fat diets increase brain GLUT2 and neuropeptide Y immunoreactivity in neonatal Wistar rats. Int J Dev Neurosci. 2010;28:625–30.CrossRefPubMed
70.
Zurück zum Zitat Yang K, Cai W, Xu J, Shi W. Maternal high-fat diet programs Wnt genes through histone modification in the liver of neonatal rats. J Mol Endocrinol. 2012;49:107–14.CrossRefPubMed Yang K, Cai W, Xu J, Shi W. Maternal high-fat diet programs Wnt genes through histone modification in the liver of neonatal rats. J Mol Endocrinol. 2012;49:107–14.CrossRefPubMed
71.
Zurück zum Zitat Tsoulis MW, Chang PE, Moore CJ, Chan KA, Gohir W, Petrik JJ, et al. Maternal high-fat diet-induced loss of fetal oocytes is associated with compromised follicle growth in adult rat offspring. Biol Reprod. 2016;94:94.CrossRefPubMedPubMedCentral Tsoulis MW, Chang PE, Moore CJ, Chan KA, Gohir W, Petrik JJ, et al. Maternal high-fat diet-induced loss of fetal oocytes is associated with compromised follicle growth in adult rat offspring. Biol Reprod. 2016;94:94.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Ebesh O, Barone A, Harper RG, Wapnir RA. Combined effect of high-fat diet and copper deficiency during gestation on fetal copper status in the rat. Biol Trace Elem Res. 1999;67:139–50.CrossRefPubMed Ebesh O, Barone A, Harper RG, Wapnir RA. Combined effect of high-fat diet and copper deficiency during gestation on fetal copper status in the rat. Biol Trace Elem Res. 1999;67:139–50.CrossRefPubMed
73.
Zurück zum Zitat Walker C-D, Naef L, d’Asti E, Long H, Xu Z, Moreau A, et al. Perinatal maternal fat intake affects metabolism and hippocampal function in the offspring a potential role for leptin. Neural Signal Oppor Nov Diagn Approaches Ther. 2008;1144:189–202. Walker C-D, Naef L, d’Asti E, Long H, Xu Z, Moreau A, et al. Perinatal maternal fat intake affects metabolism and hippocampal function in the offspring a potential role for leptin. Neural Signal Oppor Nov Diagn Approaches Ther. 2008;1144:189–202.
74.
Zurück zum Zitat Lin Y, Han X, Fang Z, Che L, Nelson J, Yan T, et al. Beneficial effects of dietary fibre supplementation of a high-fat diet on fetal development in rats. Br J Nutr. 2011;106:510–8.CrossRefPubMed Lin Y, Han X, Fang Z, Che L, Nelson J, Yan T, et al. Beneficial effects of dietary fibre supplementation of a high-fat diet on fetal development in rats. Br J Nutr. 2011;106:510–8.CrossRefPubMed
75.
Zurück zum Zitat de Oliveira AF, Fontelles CC, Rosim MP, de Oliveira TF, de Melo Loureiro AP, Mancini-Filho J, et al. Exposure to lard-based high-fat diet during fetal and lactation periods modifies breast cancer susceptibility in adulthood in rats. J Nutr Biochem. 2014;25:613–22.CrossRefPubMedPubMedCentral de Oliveira AF, Fontelles CC, Rosim MP, de Oliveira TF, de Melo Loureiro AP, Mancini-Filho J, et al. Exposure to lard-based high-fat diet during fetal and lactation periods modifies breast cancer susceptibility in adulthood in rats. J Nutr Biochem. 2014;25:613–22.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Rolls BJ, VanDuijvenvoorde PM, Rowe EA. Effects of diet and obesity on body-weight regulation during pregnancy and lactation in the rat. Physiol Behav. 1984;32:161–8.CrossRefPubMed Rolls BJ, VanDuijvenvoorde PM, Rowe EA. Effects of diet and obesity on body-weight regulation during pregnancy and lactation in the rat. Physiol Behav. 1984;32:161–8.CrossRefPubMed
77.
Zurück zum Zitat Zhang Z-Y, Dai Y-B, Wang H-N, Wang M-W. Supplementation of the maternal diet during pregnancy with chocolate and fructose interacts with the high-fat diet of the young to facilitate the onset of metabolic disorders in rat offspring. Clin Exp Pharmacol Physiol. 2013;40:652–61.CrossRefPubMed Zhang Z-Y, Dai Y-B, Wang H-N, Wang M-W. Supplementation of the maternal diet during pregnancy with chocolate and fructose interacts with the high-fat diet of the young to facilitate the onset of metabolic disorders in rat offspring. Clin Exp Pharmacol Physiol. 2013;40:652–61.CrossRefPubMed
78.
Zurück zum Zitat Cavalcante TCF, da Silva JML, da Marcelino da Silva AA, Muniz GS, da Luz Neto LM, de Souza SL, et al. Effects of a westernized diet on the reflexes and physical maturation of male rat offspring during the perinatal period. Lipids. 2013;48:1157–68.CrossRef Cavalcante TCF, da Silva JML, da Marcelino da Silva AA, Muniz GS, da Luz Neto LM, de Souza SL, et al. Effects of a westernized diet on the reflexes and physical maturation of male rat offspring during the perinatal period. Lipids. 2013;48:1157–68.CrossRef
79.
Zurück zum Zitat Cavalcante TCF, da Silva AAM, Lira MCA, do Amaral Almeida LC, Marques APJ, do Nascimento E. Early exposure of dams to a westernized diet has long-term consequences on food intake and physiometabolic homeostasis of the rat offspring. Int J Food Sci Nutr. 2014;65:989–93.CrossRef Cavalcante TCF, da Silva AAM, Lira MCA, do Amaral Almeida LC, Marques APJ, do Nascimento E. Early exposure of dams to a westernized diet has long-term consequences on food intake and physiometabolic homeostasis of the rat offspring. Int J Food Sci Nutr. 2014;65:989–93.CrossRef
80.
Zurück zum Zitat Cerf ME, Louw J, Herrera E. High fat diet exposure during fetal life enhances plasma and hepatic Omega-6 fatty acid profiles in fetal Wistar rats. Nutrients. 2015;7:7231–41.CrossRefPubMedPubMedCentral Cerf ME, Louw J, Herrera E. High fat diet exposure during fetal life enhances plasma and hepatic Omega-6 fatty acid profiles in fetal Wistar rats. Nutrients. 2015;7:7231–41.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Strakovsky RS, Zhang X, Zhou D, Pan Y-X. Gestational high fat diet programs hepatic phosphoenolpyruvate carboxykinase gene expression and histone modification in neonatal offspring rats. J Physiol. 2011;589:2707–17.CrossRefPubMedPubMedCentral Strakovsky RS, Zhang X, Zhou D, Pan Y-X. Gestational high fat diet programs hepatic phosphoenolpyruvate carboxykinase gene expression and histone modification in neonatal offspring rats. J Physiol. 2011;589:2707–17.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat de Assis S, Khan G, Hilakivi-Clarke L. High birth weight increases mammary tumorigenesis in rats. Int J Cancer. 2006;119:1537–46.CrossRefPubMed de Assis S, Khan G, Hilakivi-Clarke L. High birth weight increases mammary tumorigenesis in rats. Int J Cancer. 2006;119:1537–46.CrossRefPubMed
83.
Zurück zum Zitat Kjaergaard M, Nilsson C, Rosendal A, Nielsen MO, Raun K. Maternal chocolate and sucrose soft drink intake induces hepatic steatosis in rat offspring associated with altered lipid gene expression profile. Acta Physiol. 2014;210:142–53.CrossRef Kjaergaard M, Nilsson C, Rosendal A, Nielsen MO, Raun K. Maternal chocolate and sucrose soft drink intake induces hepatic steatosis in rat offspring associated with altered lipid gene expression profile. Acta Physiol. 2014;210:142–53.CrossRef
84.
Zurück zum Zitat Mayor RS, Finch KE, Zehr J, Morselli E, Neinast MD, Frank AP, et al. Maternal high-fat diet is associated with impaired fetal lung development. Am J Physiol Cell Mol Physiol. 2015;309:L360–8.CrossRef Mayor RS, Finch KE, Zehr J, Morselli E, Neinast MD, Frank AP, et al. Maternal high-fat diet is associated with impaired fetal lung development. Am J Physiol Cell Mol Physiol. 2015;309:L360–8.CrossRef
85.
86.
Zurück zum Zitat Bentham J, Michell AC, Lockstone H, Andrew D, Schneider JE, Brown NA, et al. Maternal high-fat diet interacts with embryonic Cited2 genotype to reduce Pitx2c expression and enhance penetrance of left-right patterning defects. Hum Mol Genet. 2010;19:3394–401.CrossRefPubMedPubMedCentral Bentham J, Michell AC, Lockstone H, Andrew D, Schneider JE, Brown NA, et al. Maternal high-fat diet interacts with embryonic Cited2 genotype to reduce Pitx2c expression and enhance penetrance of left-right patterning defects. Hum Mol Genet. 2010;19:3394–401.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat King V, Dakin RS, Liu L, Hadoke PWF, Walker BR, Seckl JR, et al. Maternal obesity has little effect on the immediate offspring but impacts on the next generation. Endocrinology. 2013;154:2514–24.CrossRefPubMed King V, Dakin RS, Liu L, Hadoke PWF, Walker BR, Seckl JR, et al. Maternal obesity has little effect on the immediate offspring but impacts on the next generation. Endocrinology. 2013;154:2514–24.CrossRefPubMed
88.
Zurück zum Zitat King V, Hibbert N, Seckl JR, Norman JE, Drake AJ. The effects of an obesogenic diet during pregnancy on fetal growth and placental gene expression are gestation dependent. Placenta. 2013;34:1087–90.CrossRefPubMed King V, Hibbert N, Seckl JR, Norman JE, Drake AJ. The effects of an obesogenic diet during pregnancy on fetal growth and placental gene expression are gestation dependent. Placenta. 2013;34:1087–90.CrossRefPubMed
89.
Zurück zum Zitat Sasson IE, Vitins AP, Mainigi MA, Moley KH, Simmons RA. Pre-gestational vs gestational exposure to maternal obesity differentially programs the offspring in mice. Diabetologia. 2015;58:615–24.CrossRefPubMed Sasson IE, Vitins AP, Mainigi MA, Moley KH, Simmons RA. Pre-gestational vs gestational exposure to maternal obesity differentially programs the offspring in mice. Diabetologia. 2015;58:615–24.CrossRefPubMed
90.
Zurück zum Zitat Edlow AG, Guedj F, Pennings JLA, Sverdlov D, Neri C, Bianchi DW. Males are from Mars, and females are from Venus: sex-specific fetal brain gene expression signatures in a mouse model of maternal diet-induced obesity. Am J Obstet Gynecol. 2016;214:623.e1–623.e10.CrossRef Edlow AG, Guedj F, Pennings JLA, Sverdlov D, Neri C, Bianchi DW. Males are from Mars, and females are from Venus: sex-specific fetal brain gene expression signatures in a mouse model of maternal diet-induced obesity. Am J Obstet Gynecol. 2016;214:623.e1–623.e10.CrossRef
91.
Zurück zum Zitat Panchenko PE, Voisin S, Jouin M, Jouneau L, Prezelin A, Lecoutre S, et al. Expression of epigenetic machinery genes is sensitive to maternal obesity and weight loss in relation to fetal growth in mice. Clin Epigenetics. 2016;8:22.CrossRefPubMedPubMedCentral Panchenko PE, Voisin S, Jouin M, Jouneau L, Prezelin A, Lecoutre S, et al. Expression of epigenetic machinery genes is sensitive to maternal obesity and weight loss in relation to fetal growth in mice. Clin Epigenetics. 2016;8:22.CrossRefPubMedPubMedCentral
92.
Zurück zum Zitat Lager S, Samulesson A-M, Taylor PD, Poston L, Powell TL, Jansson T. Diet-induced obesity in mice reduces placental efficiency and inhibits placental mTOR signaling. Physiol Rep. 2014;2:e00242.CrossRefPubMedPubMedCentral Lager S, Samulesson A-M, Taylor PD, Poston L, Powell TL, Jansson T. Diet-induced obesity in mice reduces placental efficiency and inhibits placental mTOR signaling. Physiol Rep. 2014;2:e00242.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Umekawa T, Sugiyama T, Du Q, Murabayashi N, Zhang L, Kamimoto Y, et al. A maternal mouse diet with moderately high-fat levels does not lead to maternal obesity but causes mesenteric adipose tissue dysfunction in male offspring. J Nutr Biochem. 2015;26:259–66.CrossRefPubMed Umekawa T, Sugiyama T, Du Q, Murabayashi N, Zhang L, Kamimoto Y, et al. A maternal mouse diet with moderately high-fat levels does not lead to maternal obesity but causes mesenteric adipose tissue dysfunction in male offspring. J Nutr Biochem. 2015;26:259–66.CrossRefPubMed
94.
Zurück zum Zitat Liang C, Oest ME, Jones JC, Prater MR. Gestational high saturated fat diet alters C57BL/6 mouse perinatal skeletal formation. Birth Defects Res B Dev Reprod Toxicol. 2009;86:362–9.CrossRefPubMed Liang C, Oest ME, Jones JC, Prater MR. Gestational high saturated fat diet alters C57BL/6 mouse perinatal skeletal formation. Birth Defects Res B Dev Reprod Toxicol. 2009;86:362–9.CrossRefPubMed
95.
Zurück zum Zitat Bytautiene E, Tamayo E, Kechichian T, Drever N, Gamble P, Hankins GDV, et al. Prepregnancy obesity and sFlt1-induced preeclampsia in mice: developmental programming model of metabolic syndrome. Am J Obstet Gynecol. 2011;204:398.e1–8.CrossRef Bytautiene E, Tamayo E, Kechichian T, Drever N, Gamble P, Hankins GDV, et al. Prepregnancy obesity and sFlt1-induced preeclampsia in mice: developmental programming model of metabolic syndrome. Am J Obstet Gynecol. 2011;204:398.e1–8.CrossRef
96.
Zurück zum Zitat Murabayashi N, Sugiyama T, Zhang L, Kamimoto Y, Umekawa T, Ma N, et al. Maternal high-fat diets cause insulin resistance through inflammatory changes in fetal adipose tissue. Eur J Obstet Gynecol Reprod Biol. 2013;169:39–44.CrossRefPubMed Murabayashi N, Sugiyama T, Zhang L, Kamimoto Y, Umekawa T, Ma N, et al. Maternal high-fat diets cause insulin resistance through inflammatory changes in fetal adipose tissue. Eur J Obstet Gynecol Reprod Biol. 2013;169:39–44.CrossRefPubMed
97.
Zurück zum Zitat Aye ILMH, Rosario FJ, Powell TL, Jansson T. Adiponectin supplementation in pregnant mice prevents the adverse effects of maternal obesity on placental function and fetal growth. Proc Natl Acad Sci U S A. 2015;112:12858–63.CrossRefPubMedPubMedCentral Aye ILMH, Rosario FJ, Powell TL, Jansson T. Adiponectin supplementation in pregnant mice prevents the adverse effects of maternal obesity on placental function and fetal growth. Proc Natl Acad Sci U S A. 2015;112:12858–63.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Rosario FJ, Kanai Y, Powell TL, Jansson T. Increased placental nutrient transport in a novel mouse model of maternal obesity with fetal overgrowth. Obesity. 2015;23:1663–70.CrossRefPubMed Rosario FJ, Kanai Y, Powell TL, Jansson T. Increased placental nutrient transport in a novel mouse model of maternal obesity with fetal overgrowth. Obesity. 2015;23:1663–70.CrossRefPubMed
99.
Zurück zum Zitat Masuyama H, Hiramatsu Y. Effects of a high-fat diet exposure in utero on the metabolic syndrome-like phenomenon in mouse offspring through epigenetic changes in Adipocytokine gene expression. Endocrinology. 2012;153:2823–30.CrossRefPubMed Masuyama H, Hiramatsu Y. Effects of a high-fat diet exposure in utero on the metabolic syndrome-like phenomenon in mouse offspring through epigenetic changes in Adipocytokine gene expression. Endocrinology. 2012;153:2823–30.CrossRefPubMed
100.
Zurück zum Zitat Masuyama H, Hiramatsu Y. Treatment with a constitutive androstane receptor ligand ameliorates the signs of preeclampsia in high-fat diet-induced obese pregnant mice. Mol Cell Endocrinol. 2012;348:120–7.CrossRefPubMed Masuyama H, Hiramatsu Y. Treatment with a constitutive androstane receptor ligand ameliorates the signs of preeclampsia in high-fat diet-induced obese pregnant mice. Mol Cell Endocrinol. 2012;348:120–7.CrossRefPubMed
101.
Zurück zum Zitat Dahlhoff M, Pfister S, Blutke A, Rozman J, Klingenspor M, Deutsch MJ, et al. Peri-conceptional obesogenic exposure induces sex-specific programming of disease susceptibilities in adult mouse offspring. Biochim Biophys Acta. 1842;2014:304–17. Dahlhoff M, Pfister S, Blutke A, Rozman J, Klingenspor M, Deutsch MJ, et al. Peri-conceptional obesogenic exposure induces sex-specific programming of disease susceptibilities in adult mouse offspring. Biochim Biophys Acta. 1842;2014:304–17.
103.
Zurück zum Zitat Masuyama H, Mitsui T, Nobumoto E, Hiramatsu Y. The effects of high-fat diet exposure in utero on the obesogenic and Diabetogenic traits through epigenetic changes in adiponectin and leptin gene expression for multiple generations in female mice. Endocrinology. 2015;156:2482–91.CrossRefPubMed Masuyama H, Mitsui T, Nobumoto E, Hiramatsu Y. The effects of high-fat diet exposure in utero on the obesogenic and Diabetogenic traits through epigenetic changes in adiponectin and leptin gene expression for multiple generations in female mice. Endocrinology. 2015;156:2482–91.CrossRefPubMed
104.
Zurück zum Zitat Sferruzzi-Perri AN, Vaughan OR, Haro M, Cooper WN, Musial B, Charalambous M, et al. An obesogenic diet during mouse pregnancy modifies maternal nutrient partitioning and the fetal growth trajectory. FASEB J. 2013;27:3928–37.CrossRefPubMed Sferruzzi-Perri AN, Vaughan OR, Haro M, Cooper WN, Musial B, Charalambous M, et al. An obesogenic diet during mouse pregnancy modifies maternal nutrient partitioning and the fetal growth trajectory. FASEB J. 2013;27:3928–37.CrossRefPubMed
105.
Zurück zum Zitat Turdi S, Ge W, Hu N, Bradley KM, Wang X, Ren J. Interaction between maternal and postnatal high fat diet leads to a greater risk of myocardial dysfunction in offspring via enhanced lipotoxicity, IRS-1 serine phosphorylation and mitochondrial defects. J Mol Cell Cardiol. 2013;55:117–29.CrossRefPubMed Turdi S, Ge W, Hu N, Bradley KM, Wang X, Ren J. Interaction between maternal and postnatal high fat diet leads to a greater risk of myocardial dysfunction in offspring via enhanced lipotoxicity, IRS-1 serine phosphorylation and mitochondrial defects. J Mol Cell Cardiol. 2013;55:117–29.CrossRefPubMed
106.
Zurück zum Zitat del Mar PM, Williams L, Seki Y, Hartil K, Kaur H, Lin C-L, et al. Critical periods of increased fetal vulnerability to a maternal high fat diet. Reprod Biol Endocrinol. 2014;12:80.CrossRef del Mar PM, Williams L, Seki Y, Hartil K, Kaur H, Lin C-L, et al. Critical periods of increased fetal vulnerability to a maternal high fat diet. Reprod Biol Endocrinol. 2014;12:80.CrossRef
107.
Zurück zum Zitat Luijten M, Singh AV, Bastian CA, Westerman A, Pisano MM, Pennings JLA, et al. Lasting effects on body weight and mammary gland gene expression in female mice upon early life exposure to n-3 but not n-6 high-fat diets. PLoS One. 2013;8:e55603.CrossRefPubMedPubMedCentral Luijten M, Singh AV, Bastian CA, Westerman A, Pisano MM, Pennings JLA, et al. Lasting effects on body weight and mammary gland gene expression in female mice upon early life exposure to n-3 but not n-6 high-fat diets. PLoS One. 2013;8:e55603.CrossRefPubMedPubMedCentral
108.
Zurück zum Zitat Benatti RO, Melo AM, Borges FO, Ignacio-Souza LM, Simino LAP, Milanski M, et al. Maternal high-fat diet consumption modulates hepatic lipid metabolism and microRNA-122 (miR-122) and microRNA-370 (miR-370) expression in offspring. Br J Nutr. 2014;111:2112–22.CrossRefPubMed Benatti RO, Melo AM, Borges FO, Ignacio-Souza LM, Simino LAP, Milanski M, et al. Maternal high-fat diet consumption modulates hepatic lipid metabolism and microRNA-122 (miR-122) and microRNA-370 (miR-370) expression in offspring. Br J Nutr. 2014;111:2112–22.CrossRefPubMed
109.
Zurück zum Zitat Volpato AM, Schultz A, Magalhaes-da-Costa E, de Gusmao Correia ML, Aguila MB, Mandarim-de-Lacerda CA. Maternal high-fat diet programs for metabolic disturbances in offspring despite leptin sensitivity. Neuroendocrinology. 2012;96:272–84.CrossRefPubMed Volpato AM, Schultz A, Magalhaes-da-Costa E, de Gusmao Correia ML, Aguila MB, Mandarim-de-Lacerda CA. Maternal high-fat diet programs for metabolic disturbances in offspring despite leptin sensitivity. Neuroendocrinology. 2012;96:272–84.CrossRefPubMed
110.
Zurück zum Zitat Ashino NG, Saito KN, Souza FD, Nakutz FS, Roman EA, Velloso LA, et al. Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J Nutr Biochem. 2012;23:341–8 Elsevier Inc.CrossRefPubMed Ashino NG, Saito KN, Souza FD, Nakutz FS, Roman EA, Velloso LA, et al. Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J Nutr Biochem. 2012;23:341–8 Elsevier Inc.CrossRefPubMed
111.
Zurück zum Zitat Gregorio BM, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Maternal high-fat diet is associated with altered pancreatic remodelling in mice offspring. Eur J Nutr. 2013;52:759–69.CrossRefPubMed Gregorio BM, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Maternal high-fat diet is associated with altered pancreatic remodelling in mice offspring. Eur J Nutr. 2013;52:759–69.CrossRefPubMed
112.
Zurück zum Zitat Chin EH, Christians JK. When are sex-specific effects really sex-specific? J Dev Orig Health Dis. 2015;6:438–42.CrossRefPubMed Chin EH, Christians JK. When are sex-specific effects really sex-specific? J Dev Orig Health Dis. 2015;6:438–42.CrossRefPubMed
113.
Zurück zum Zitat Dodson RB, Miller TA, Powers K, Yang Y, Yu B, Albertine KH, et al. Intrauterine growth restriction influences vascular remodeling and stiffening in the weanling rat more than sex or diet. Am J Physiol Heart Circ Physiol. 2017;312:H250–64.CrossRefPubMed Dodson RB, Miller TA, Powers K, Yang Y, Yu B, Albertine KH, et al. Intrauterine growth restriction influences vascular remodeling and stiffening in the weanling rat more than sex or diet. Am J Physiol Heart Circ Physiol. 2017;312:H250–64.CrossRefPubMed
114.
Zurück zum Zitat Huang Y, Ye T, Liu C, Fang F, Chen Y, Dong Y. Maternal high-fat diet during pregnancy and lactation affects hepatic lipid metabolism in early life of offspring rat. J Biosci. 2017;42:311–9.CrossRefPubMed Huang Y, Ye T, Liu C, Fang F, Chen Y, Dong Y. Maternal high-fat diet during pregnancy and lactation affects hepatic lipid metabolism in early life of offspring rat. J Biosci. 2017;42:311–9.CrossRefPubMed
115.
Zurück zum Zitat Ye K, Li L, Zhang D, Li Y, Wang HQ, Lai HL, et al. Effect of maternal obesity on fetal growth and expression of placental fatty acid transporters. J Clin Res Pediatr Endocrinol. 2017;9:300–7.PubMedPubMedCentral Ye K, Li L, Zhang D, Li Y, Wang HQ, Lai HL, et al. Effect of maternal obesity on fetal growth and expression of placental fatty acid transporters. J Clin Res Pediatr Endocrinol. 2017;9:300–7.PubMedPubMedCentral
116.
Zurück zum Zitat Yamada-Obara N, Yamagishi S, Taguchi K, Kaida Y, Yokoro M, Nakayama Y, et al. Maternal exposure to high-fat and high-fructose diet evokes hypoadiponectinemia and kidney injury in rat offspring. Clin Exp Nephrol. 2016;20:853–61.CrossRefPubMed Yamada-Obara N, Yamagishi S, Taguchi K, Kaida Y, Yokoro M, Nakayama Y, et al. Maternal exposure to high-fat and high-fructose diet evokes hypoadiponectinemia and kidney injury in rat offspring. Clin Exp Nephrol. 2016;20:853–61.CrossRefPubMed
117.
Zurück zum Zitat Lecoutre S, Deracinois B, Laborie C, Eberlé D, Guinez C, Panchenko PE, et al. Depot- and sex-specific effects of maternal obesity in offspring’s adipose tissue. J Endocrinol. 2016;230:39–53.CrossRefPubMed Lecoutre S, Deracinois B, Laborie C, Eberlé D, Guinez C, Panchenko PE, et al. Depot- and sex-specific effects of maternal obesity in offspring’s adipose tissue. J Endocrinol. 2016;230:39–53.CrossRefPubMed
118.
Zurück zum Zitat Bae-Gartz I, Janoschek R, Kloppe CS, Vohlen C, Roels F, Oberthür A, et al. Running exercise in obese pregnancies prevents IL-6 trans-signaling in male offspring. Med Sci Sports Exerc. 2016;48:829–38.CrossRefPubMed Bae-Gartz I, Janoschek R, Kloppe CS, Vohlen C, Roels F, Oberthür A, et al. Running exercise in obese pregnancies prevents IL-6 trans-signaling in male offspring. Med Sci Sports Exerc. 2016;48:829–38.CrossRefPubMed
119.
Zurück zum Zitat Chin EH, Schmidt KL, Martel KM, Wong CK, Hamden JE, Gibson WT, et al. A maternal high-fat, high-sucrose diet has sex-specific effects on fetal glucocorticoids with little consequence for offspring metabolism and voluntary locomotor activity in mice. PLoS One. 2017;12:e0174030.CrossRefPubMedPubMedCentral Chin EH, Schmidt KL, Martel KM, Wong CK, Hamden JE, Gibson WT, et al. A maternal high-fat, high-sucrose diet has sex-specific effects on fetal glucocorticoids with little consequence for offspring metabolism and voluntary locomotor activity in mice. PLoS One. 2017;12:e0174030.CrossRefPubMedPubMedCentral
120.
Zurück zum Zitat Jonscher KR, Stewart MS, Alfonso-Garcia A, DeFelice BC, Wang XX, Luo Y, et al. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice. FASEB J. 2017;31:1434–48.CrossRefPubMed Jonscher KR, Stewart MS, Alfonso-Garcia A, DeFelice BC, Wang XX, Luo Y, et al. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice. FASEB J. 2017;31:1434–48.CrossRefPubMed
121.
Zurück zum Zitat Connor KL, Chehoud C, Altrichter A, Chan L, DeSantis TZ, Lye SJ. Maternal metabolic, immune, and microbial systems in late pregnancy vary with malnutrition in mice. Biol Reprod. 2018;98:579–92.PubMed Connor KL, Chehoud C, Altrichter A, Chan L, DeSantis TZ, Lye SJ. Maternal metabolic, immune, and microbial systems in late pregnancy vary with malnutrition in mice. Biol Reprod. 2018;98:579–92.PubMed
122.
Zurück zum Zitat Masuyama H, Mitsui T, Maki J, Tani K, Nakamura K, Hiramatsu Y. Dimethylesculetin ameliorates maternal glucose intolerance and fetal overgrowth in high-fat diet-fed pregnant mice via constitutive androstane receptor. Mol Cell Biochem. 2016;419:185–92.CrossRefPubMed Masuyama H, Mitsui T, Maki J, Tani K, Nakamura K, Hiramatsu Y. Dimethylesculetin ameliorates maternal glucose intolerance and fetal overgrowth in high-fat diet-fed pregnant mice via constitutive androstane receptor. Mol Cell Biochem. 2016;419:185–92.CrossRefPubMed
123.
Zurück zum Zitat Nam J, Greenwald E, Jack-Roberts C, Ajeeb TT, Malysheva OV, Caudill MA, et al. Choline prevents fetal overgrowth and normalizes placental fatty acid and glucose metabolism in a mouse model of maternal obesity. J Nutr Biochem. 2017;49:80–8.CrossRefPubMedPubMedCentral Nam J, Greenwald E, Jack-Roberts C, Ajeeb TT, Malysheva OV, Caudill MA, et al. Choline prevents fetal overgrowth and normalizes placental fatty acid and glucose metabolism in a mouse model of maternal obesity. J Nutr Biochem. 2017;49:80–8.CrossRefPubMedPubMedCentral
124.
Zurück zum Zitat Lai M, Chandrasekera PC, Barnard ND. You are what you eat, or are you? The challenges of translating high-fat-fed rodents to human obesity and diabetes. Nutr Diabetes. 2014;4:e135. Lai M, Chandrasekera PC, Barnard ND. You are what you eat, or are you? The challenges of translating high-fat-fed rodents to human obesity and diabetes. Nutr Diabetes. 2014;4:e135.
125.
Zurück zum Zitat Pellizzon MA, Ricci MR. The common use of improper control diets in diet-induced metabolic disease research confounds data interpretation: The fiber factor. Nutr Metab. 2018;15:3. Pellizzon MA, Ricci MR. The common use of improper control diets in diet-induced metabolic disease research confounds data interpretation: The fiber factor. Nutr Metab. 2018;15:3.
126.
Zurück zum Zitat Austin GL, Ogden LG, Hill JO. Trends in carbohydrate, fat, and protein intakes and association with energy intake in normal-weight, overweight, and obese individuals: 1971-2006. Am J Clin Nutr. 2011;93:836–43.CrossRefPubMed Austin GL, Ogden LG, Hill JO. Trends in carbohydrate, fat, and protein intakes and association with energy intake in normal-weight, overweight, and obese individuals: 1971-2006. Am J Clin Nutr. 2011;93:836–43.CrossRefPubMed
127.
Zurück zum Zitat Ford ES, Dietz WH. Trends in energy intake among adults in the United States: findings from NHANES. Am J Clin Nutr. 2013;97:848–53.CrossRefPubMed Ford ES, Dietz WH. Trends in energy intake among adults in the United States: findings from NHANES. Am J Clin Nutr. 2013;97:848–53.CrossRefPubMed
128.
Zurück zum Zitat Wright JD, Wang C, Kennedy-Stephenson J, Ervin RB. Dietary intake of ten key nutrients for public health, United States: 1999-2000. Adv Data. 2003;334:1–4. Wright JD, Wang C, Kennedy-Stephenson J, Ervin RB. Dietary intake of ten key nutrients for public health, United States: 1999-2000. Adv Data. 2003;334:1–4.
129.
Zurück zum Zitat Frihauf JB, Fekete ÉM, Nagy TR, Levin BE, Zorrilla EP. Maternal Western diet increases adiposity even in male offspring of obesity-resistant rat dams: early endocrine risk markers. Am J Physiol Regul Integr Comp Physiol. 2016;311:R1045–59.CrossRefPubMedPubMedCentral Frihauf JB, Fekete ÉM, Nagy TR, Levin BE, Zorrilla EP. Maternal Western diet increases adiposity even in male offspring of obesity-resistant rat dams: early endocrine risk markers. Am J Physiol Regul Integr Comp Physiol. 2016;311:R1045–59.CrossRefPubMedPubMedCentral
130.
Zurück zum Zitat Shankar K, Harrell A, Liu X, Gilchrist JM, Ronis MJJ, Badger TM. Maternal obesity at conception programs obesity in the offspring. Am J Physiol Regul Integr Comp Physiol. 2008;294:R528–38.CrossRefPubMed Shankar K, Harrell A, Liu X, Gilchrist JM, Ronis MJJ, Badger TM. Maternal obesity at conception programs obesity in the offspring. Am J Physiol Regul Integr Comp Physiol. 2008;294:R528–38.CrossRefPubMed
131.
Zurück zum Zitat Lin Y, Zhuo Y, Fang Z, Che L, Wu D. Effect of maternal dietary energy types on placenta nutrient transporter gene expressions and intrauterine fetal growth in rats. Nutrition. 2012;28:1037–43.CrossRefPubMed Lin Y, Zhuo Y, Fang Z, Che L, Wu D. Effect of maternal dietary energy types on placenta nutrient transporter gene expressions and intrauterine fetal growth in rats. Nutrition. 2012;28:1037–43.CrossRefPubMed
132.
Zurück zum Zitat Fornes R, Maliqueo M, Hu M, Hadi L, Jimenez-Andrade JM, Ebefors K, et al. The effect of androgen excess on maternal metabolism, placental function and fetal growth in obese dams. Sci Rep. 2017;7:8066.CrossRefPubMedPubMedCentral Fornes R, Maliqueo M, Hu M, Hadi L, Jimenez-Andrade JM, Ebefors K, et al. The effect of androgen excess on maternal metabolism, placental function and fetal growth in obese dams. Sci Rep. 2017;7:8066.CrossRefPubMedPubMedCentral
133.
Zurück zum Zitat Djelantik AAAMJ, Kunst AE, Van Der Wal MF, Smit HA, Vrijkotte TGM. Contribution of overweight and obesity to the occurrence of adverse pregnancy outcomes in a multi-ethnic cohort: population attributive fractions for Amsterdam. BJOG An Int J Obstet Gynaecol. 2012;119:283–90.CrossRef Djelantik AAAMJ, Kunst AE, Van Der Wal MF, Smit HA, Vrijkotte TGM. Contribution of overweight and obesity to the occurrence of adverse pregnancy outcomes in a multi-ethnic cohort: population attributive fractions for Amsterdam. BJOG An Int J Obstet Gynaecol. 2012;119:283–90.CrossRef
134.
Zurück zum Zitat Li N, Liu E, Guo J, Pan L, Li B, Wang P, et al. Maternal prepregnancy body mass index and gestational weight gain on pregnancy outcomes. PLoS One. 2013;8:e82310.CrossRefPubMedPubMedCentral Li N, Liu E, Guo J, Pan L, Li B, Wang P, et al. Maternal prepregnancy body mass index and gestational weight gain on pregnancy outcomes. PLoS One. 2013;8:e82310.CrossRefPubMedPubMedCentral
135.
Zurück zum Zitat Scott-Pillai R, Spence D, Cardwell CR, Hunter A, Holmes VA. The impact of body mass index on maternal and neonatal outcomes: a retrospective study in a UK obstetric population, 2004-2011. Br J Obstet Gynaecol. 2013;120:932–9.CrossRef Scott-Pillai R, Spence D, Cardwell CR, Hunter A, Holmes VA. The impact of body mass index on maternal and neonatal outcomes: a retrospective study in a UK obstetric population, 2004-2011. Br J Obstet Gynaecol. 2013;120:932–9.CrossRef
136.
Zurück zum Zitat Sebire NJ, Jolly M, Harris JP, Wadsworth J, Joffe M, Beard RW, et al. Maternal obesity and pregnancy outcome: a study of 287 213 pregnancies in London. Int J Obes. 2001;25:1175–82.CrossRef Sebire NJ, Jolly M, Harris JP, Wadsworth J, Joffe M, Beard RW, et al. Maternal obesity and pregnancy outcome: a study of 287 213 pregnancies in London. Int J Obes. 2001;25:1175–82.CrossRef
137.
138.
Zurück zum Zitat Villar J, Carroli G, Wojdyla D, Abalos E, Giordano D, Ba’aqeel H, et al. Preeclampsia, gestational hypertension and intrauterine growth restriction, related or independent conditions? Am J Obstet Gynecol. 2006;194:921–31.CrossRefPubMed Villar J, Carroli G, Wojdyla D, Abalos E, Giordano D, Ba’aqeel H, et al. Preeclampsia, gestational hypertension and intrauterine growth restriction, related or independent conditions? Am J Obstet Gynecol. 2006;194:921–31.CrossRefPubMed
139.
Zurück zum Zitat Weiss JL, Malone FD, Emig D, Ball RH, Nyberg DA, Comstock CH, et al. Obesity, obstetric complications and cesarean delivery rate - a population-based screening study. Am J Obstet Gynecol. 2004;190:1091–7.CrossRefPubMed Weiss JL, Malone FD, Emig D, Ball RH, Nyberg DA, Comstock CH, et al. Obesity, obstetric complications and cesarean delivery rate - a population-based screening study. Am J Obstet Gynecol. 2004;190:1091–7.CrossRefPubMed
140.
Zurück zum Zitat Cedergren MI. Maternal morbid obesity and the risk of adverse pregnancy outcome. Obstet Gynecol. 2004;103:219–24.CrossRefPubMed Cedergren MI. Maternal morbid obesity and the risk of adverse pregnancy outcome. Obstet Gynecol. 2004;103:219–24.CrossRefPubMed
141.
Zurück zum Zitat Reynolds RM, Allan KM, Raja EA, Bhattacharya S, McNeill G, Hannaford PC, et al. Maternal obesity during pregnancy and premature mortality from cardiovascular events in adult offspring: follow-up of 1,323,275 person years. BMJ. 2013;347:f4539.CrossRefPubMedPubMedCentral Reynolds RM, Allan KM, Raja EA, Bhattacharya S, McNeill G, Hannaford PC, et al. Maternal obesity during pregnancy and premature mortality from cardiovascular events in adult offspring: follow-up of 1,323,275 person years. BMJ. 2013;347:f4539.CrossRefPubMedPubMedCentral
142.
Zurück zum Zitat Dickinson H, Moss TJ, Gatford KL, Moritz KM, Akison L, Fullston T, et al. A review of fundamental principles for animal models of DOHaD research: an Australian perspective. J Dev Orig Health Dis. 2016;7:449–72.CrossRefPubMed Dickinson H, Moss TJ, Gatford KL, Moritz KM, Akison L, Fullston T, et al. A review of fundamental principles for animal models of DOHaD research: an Australian perspective. J Dev Orig Health Dis. 2016;7:449–72.CrossRefPubMed
143.
Zurück zum Zitat Williams L, Seki Y, Vuguin PM, Charron MJ. Animal models of in utero exposure to a high fat diet: a review. Biochim Biophys Acta-Molecular Basis Dis. 1842;2014:507–19. Williams L, Seki Y, Vuguin PM, Charron MJ. Animal models of in utero exposure to a high fat diet: a review. Biochim Biophys Acta-Molecular Basis Dis. 1842;2014:507–19.
144.
Zurück zum Zitat Montgomery MK, Hallahan NL, Brown SH, Liu M, Mitchell TW, Cooney GJ, et al. Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia. 2013;56:1129–39.CrossRefPubMed Montgomery MK, Hallahan NL, Brown SH, Liu M, Mitchell TW, Cooney GJ, et al. Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia. 2013;56:1129–39.CrossRefPubMed
145.
Zurück zum Zitat Simon MM, Greenaway S, White JK, Fuchs H, Gailus-Durner V, Wells S, et al. A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains. Genome Biol. 2013;14:R82.CrossRefPubMedPubMedCentral Simon MM, Greenaway S, White JK, Fuchs H, Gailus-Durner V, Wells S, et al. A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains. Genome Biol. 2013;14:R82.CrossRefPubMedPubMedCentral
146.
Zurück zum Zitat Morrison JL, Botting KJ, Darby JRT, David AL, Dyson RM, Gatford KL, et al. Guinea pig models for translation of the developmental origins of health and disease hypothesis into the clinic. J Physiol. 2018;596:5535–69.CrossRefPubMedPubMedCentral Morrison JL, Botting KJ, Darby JRT, David AL, Dyson RM, Gatford KL, et al. Guinea pig models for translation of the developmental origins of health and disease hypothesis into the clinic. J Physiol. 2018;596:5535–69.CrossRefPubMedPubMedCentral
147.
Zurück zum Zitat Chavatte-Palmer P, Tarrade A, Rousseau-Ralliard D. Diet before and during pregnancy and offspring health: the importance of animal models and what can be learned from them. Int J Environ Res Public Health. 2016;13:586.CrossRefPubMedCentral Chavatte-Palmer P, Tarrade A, Rousseau-Ralliard D. Diet before and during pregnancy and offspring health: the importance of animal models and what can be learned from them. Int J Environ Res Public Health. 2016;13:586.CrossRefPubMedCentral
Metadaten
Titel
Effects of high-fat diets on fetal growth in rodents: a systematic review
verfasst von
Julian K. Christians
Kendra I. Lennie
Lisa K. Wild
Raajan Garcha
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2019
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-019-0482-y

Weitere Artikel der Ausgabe 1/2019

Reproductive Biology and Endocrinology 1/2019 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.