Skip to main content
Erschienen in: Journal of Translational Medicine 1/2015

Open Access 01.12.2015 | Review

Milk consumption during pregnancy increases birth weight, a risk factor for the development of diseases of civilization

verfasst von: Bodo C Melnik, Swen Malte John, Gerd Schmitz

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2015

Abstract

Antenatal dietary lifestyle intervention and nutrition during pregnancy and early postnatal life are important for appropriate lifelong metabolic programming. Epidemiological evidence underlines the crucial role of increased birth weight as a risk factor for the development of chronic diseases of civilization such as obesity, diabetes and cancer. Obstetricians and general practitioners usually recommend milk consumption during pregnancy as a nutrient enriched in valuable proteins and calcium for bone growth. However, milk is not just a simple nutrient, but has been recognized to function as an endocrine signaling system promoting anabolism and postnatal growth by activating the nutrient-sensitive kinase mTORC1. Moreover, pasteurized cow’s milk transfers biologically active exosomal microRNAs into the systemic circulation of the milk consumer apparently affecting more than 11 000 human genes including the mTORC1-signaling pathway. This review provides literature evidence and evidence derived from translational research that milk consumption during pregnancy increases gestational, placental, fetal and birth weight. Increased birth weight is a risk factor for the development of diseases of civilization thus involving key disciplines of medicine. With regard to the presented evidence we suggest that dietary recommendations promoting milk consumption during pregnancy have to be re-evaluated.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

BCM was responsible for translational research and data abstraction. All authors performed and contributed to literature search, reviewed and edited the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AAs
Amino acids
AGA
Appropriate for gestational age
BCAA
Branched-chain amino acid
BM
Basal membrane
BMI
Body mass index
4E-BP-1
Eukaryotic translation initiation factor 4E-binding protein 1
FGF21
Fibroblast growth factor 21
GDH
Glutamate dehydrogenase
GDM
Gestational diabetes mellitus
GHR
Growth hormone receptor
GIP
Glucose-dependent insulinotropic polypeptide
GLP-1
Glucagon-like peptide-1
HPL
Human placental lactogen
IGF-1
Insulin-like growth factor-1
IGF1R
Insulin-like growth factor-1 receptor
IR
Insulin receptor
IRS
Insulin receptor substrate
JAK
Janus-activated kinase
PI3K
Phosphoinositide-3 kinase
LAT
L-type amino acid transporter
Leu
Leucine
LGA
Large for gestational age
MicroRNA
Micro-ribonucleic acid
mTORC1
Mechanistic target of rapamycin complex 1
MVM
Microvillous plasma membrane
PGH
Placental growth hormone
PRLR
Prolactin receptor
PTEN
Phosphatase and tensin homolog
RHEB
RAS homolog enrich in brain
S6K1
Ribosomal protein S6 kinase, 70-kD kinase 1
SOCS
Suppressor of cytokine signaling
TSC2
Tuberin

Introduction

To meet the requirements for macronutrients and minerals during pregnancy gynecologists and general practitioners recommend increased servings of milk and dairy products [1]. Although milk is a rich source of essential amino acids and calcium, recent understanding of milk’s biological function has changed during the last years. Milk is not just a nutrient, but represents an endocrine signaling system of mammals activating the key regulator of cell growth and anabolism, the nutrient-sensitive kinase mTORC1 (mechanistic target of rapamycin complex 1) [2]. At the molecular level, cell growth, proliferation, and anabolism are regulated by mTORC1 [3-12]. In the perspective of human evolution, persistent cow’s milk consumption is a novel human behavior, which may result in long-term adverse health effects [13]. Increased mTORC1 signaling during milk consumption has recently been confirmed in a rodent model and has been associated with the development of obesity [14-17]. Pregravid maternal overweight and obesity are well-known risk factors that promote fetal overnutrition and fetal macrosomia [18-26]. Increased birth weight is a risk factor for the development of diseases of civilization, especially obesity [22,23,25]. To understand the impact of milk consumption on fetal growth, it is of critical importance to appreciate milk’s biological function as an activator of mTORC1 and transmitter of gene-regulatory exosomal microRNAs [2].

Pivotal mTORC1-activating signals

mTORC1 orchestrates cell growth and proliferation [3]. mTORC1 is the central hub of metabolism that activates nucleotide, protein and lipid synthesis under conditions of nutrient and growth factor availability [3-12]. mTORC1 plays a fundamental role in cell cycle control and cell growth [27], protein and lipid synthesis [7,12], lipid accumulation and adipogenesis [28,29]. Thus, persistently overactivated mTORC1 signaling stimulates weight gain, increases body mass, and fat mass [14,29,30].
Basically, there are five major pathways, that activate mTORC1: 1) the presence of growth factors such as insulin and IGF-1 [3,6,7,11,12], 2) sufficient cellular energy (glucose, ATP) [31,32], 3) the availability of amino acids, predominantly essential branched-chain amino acids (BCAAs) such as leucine [5-10,33], 4) the presence of glutamine for cellular leucine uptake and glutaminolysis-mediated activation of mTORC1 [34-36], and 5) the availability of saturated fatty acids, especially palmitic acid [37].

Milk provides all signals for mTORC1-activation

Mammalian evolution relies on lactation and its secretory end-product milk, required and sufficient for postnatal growth. Milk is not just a simple nutrient, but represents a sophisticated postnatal endocrine system providing all signals that are required to activate mTORC1 of the milk recipient, physiologically the newborn mammal [2].

Essential branched-chain amino acids activate mTORC1

Milk proteins provide highest amounts of essential BCAAs, especially leucine [38]. Leucine plays a pivotal role for activating mTORC1 (Figure 1) [33]. Of all animal proteins, whey proteins contain the highest amount of leucine (14%) [38], and in comparsion to meat (8% leucine), whey proteins undergo fast intestinal hydrolysis, thus operate like an i.v. amino acid infusion [39-42].

Glutamine activates mTORC1

Milk protein (8.09 g glutamine/100 g) in comparison to beef protein (4.75 g glutamine/100 g) provides 70% more glutamine [43]. Glutamine is an important activator of mTORC1 via its function as a gatekeeper for cellular leucine uptake and via its precursor function in the glutaminolysis pathway that activates mTORC1 (Figure 1) [44-46]. Leucine is an allosteric activator of glutamate dehydrogenase (GDH), the key-regulating enzyme of the glutaminolysis pathway [44,45]. The interplay of glutamine and leucine maximizes the flux through GDH in pancreatic β-cells, which is important for mTORC1-S6K1-dependent insulin secretion [46].

Insulin activates mTORC1

Milk stimulates insulin secretion (Figure 1) [2]. The insulinemic index of whole cow’s milk (148 ± 14) and skim milk (140 ± 13) is much higher than the glycemic indices of whole milk (42 ± 5) and skim milk (37 ± 9), respectively [47,48]. Fast hydrolysis and immediate intestinal absorption of insulinotropic amino acids of the whey protein fraction of cow’s milk raises insulin levels to much higher magnitudes than intestinal digestion of structural proteins such as beef (insulinemic index: 51) [47,48]. The major insulinotropic protein fraction of cow’s milk is the whey protein fraction [49]. Whey-derived leucine and other whey-derived amino acids stimulate incretin secretion of enteroendocrine K- and L-cells [50-54]. Additionally, whey-derived amino acids directly exert inulinotropic effects on pancreatic β-cells [55-57]. Milk protein consumption in comparison to meat protein intake thus results in hyperinsulinemia [58].

Insulin-like growth factor-1 activates mTORC1

A meta-analysis confirmed that continued milk consumption increases serum levels of insulin-like growth factor-1 (IGF-1) [59]. The European Prospective Investigation into Cancer and Nutrition confirmed a relationship between milk intake in 2 109 European women with increased IGF-1 serum levels [60]. A 20% increase in serum IGF-1 levels has been observed in prepubertal children previously not used to milk consumption after a daily intake of 710 mL of milk for 4 weeks [61]. A recent study including 193 overweight adolescents aged 12–15 years drank either 1 L/day of skimmed milk, whey, casein or water for 12 weeks. All milk-based-drinks contained 35 g milk protein/L. IGF-1 significantly increased with skimmed milk and tended to increase with casein compared to the pre-test control group [62]. Casein in comparison to whey protein has been shown to differentially enhance hepatic IGF-1 synthesis [49]. Notably, per capita cheese consumption, the major dairy source of casein, increased in Germany from 5 kg in 1950 to 24.4 kg in 2013 [63].

Palmitic acid activates mTORC1

Cow’s milk contains about 3.5 to 5% total lipid. About 98% of the lipid is composed of triacylglycerol, transported in milk fat globules [64]. The major fatty acid of total fatty acids of milk lipids is palmitate (C16:0) with 32.3 wt% [64,65]. Palmitate like BCAAs activates mTORC1 [37].
Thus milk, the promoter of postnatal growth of mammals, activates mTORC1 of the milk recipient either by transfer or induction of critical mTORC1 activating signals (Figure 1).
It is the intention of this review to demonstrate that milk consumption during pregnancy increases weight trajectories of the growing human fetus promoting increased birth weight, a well-known risk factor for the development of diseases of civilization.

Milk consumption and pregravid maternal weight

Prepregnancy maternal overweight and obesity are risk factors promoting fetal overnutrition and macrosomia [18-26]. Obesity is associated with enhanced TORC1 signaling [14-16]. In obesity serum levels of insulin, BCAAs, and free palmitate are increased [66-69]. In obese children additional supply of leucine resulted in excessive hyperinsulinemia [70]. Elevated serum levels of BCAAs in children and adolescents have been identified as predictors of insulin resistance [69]. Notably, milk protein but not meat protein consumption induced hyperinsulinemia and insulin resistance [58]. In obesity and states of insulin resistance, palmitate serum levels are significantly elevated [71-73]. Milk-mediated stimulation of mTORC1 increases the phosphorylation of the major mTORC1 substrate, S6 kinase 1 (S6K1) [14]. Overactivated S6K1 via phosphorylation of insulin receptor substrate-1 (IRS-1) is a pivotal mechanism that induces insulin resistance [74,75].
There is substantial evidence that milk consumption in children increases linear growth and body mass index (BMI) [76-78], increases BMI in adolescents, and adults [79-81]. Noteworthy, a recent meta-analysis of Chen et al. [82] including 29 randomized controlled trials found no significant effects of total dairy intake on body weight and body fat [82]. Notably, this study did not differentiate between milk and other processed milk products. The study of Abreu et al. [83] reported a protective association between dairy product consumption and abdominal obesity among Azorean boys. However, this study using a self-administered semiquantitative food frequency questionnaire (categorizing < 2 and ≥ 2 servings per day) did not discriminate between the effects of milk consumption versus other dairy products and did not provide quantitative dose–response data on daily milk intake. By using the same semiquantitative food frequency questionnaire methodology categorizing the number of servings/day the authors reported an inverse association between milk intake and both BMI and body fat in 583 Azorean girls but not in 418 Azorean boys [84]. In contrast, Arnberg et al. [80] investigated 203 overweight adolescents with a BMI of 25.4 ± 2.3 kg/m2 (mean ± SD), who received an additional daily amount of 35 g milk protein either as 1 L/day of skim milk, whey, or casein, or water as a control for 12 weeks. BMI-for-age Z-score was greater at 12 weeks in the skim milk, whey, and casein groups compared with baseline and the control groups [80]. Remarkably, the National Health and Nutrition Examination Survey (NHANES) data from 1999 to 2004 including 1,493 children of age 2–4 years and 2,526 children of age 5–10 years reported an association for the highest quartiles of milk consumption and BMI in contrast to other dairy products, which had no effect on BMI [77]. It is of critical concern that increased prepregnancy BMI represents an important risk factor for increased birth weight of the offspring [21,22,25].

Milk consumption and gestational weight gain

Women with large for gestational age (LGA) newborns had an increased BMI before pregnancy (25 kg/m2), an increased gestational weight gain of 19.0 kg in comparison to women with a normal BMI before pregnancy of 22.4 kg/m2 exhibiting a gestational weight gain of 15.8 kg, respectively [85]. Intriguingly, pregnant women gaining excessive weight in comparison to women with optimal weight gain reported a twofold intake of dairy products of about 200 g/day [86]. Of all dairy products, the strongest predictor of increased maternal weight gain during the last trimester of pregnancy was milk [86]. Thus, milk consumption during pregnancy may increase gestational weight gain.

Milk consumption and placental weight

Data from 50 117 mother-infant pairs of the Danish National Birth Cohort collected from 1996–2002 showed a placental weight increase across the whole range of milk intake [87]. A linear increment of placental weight from 13.3 g (0–1 glass of milk/day) to 26.4 g (>6 glasses of milk/day) (p < 0.001) has been reported [87]. A prospective study in India reported that the frequency of milk consumption at 18th week of gestation was positively associated with an increase of placental weight [88].
A milk-induced increase in placental weight may not only raise the nutrient transfer to the fetus but may also increase the amount of placenta-derived growth hormones that impair maternal insulin sensitivity, thereby enhancing maternal blood glucose levels leading to fetal overgrowth and increased birth weight. In fact, maternal blood levels of human placental lactogen (HPL) are correlated with placental weight [89-91], and fetal weight [91-94], respectively. CSH1, the predominant transcript of HPL, is increased in placentas of LGA pregnancies [95]. A link between fetal growth velocity in the second half of the pregnancy and maternal serum HPL levels has been demonstrated [96]. In LGA newborns the expression of CSH1-1, CSH2-1, and CSHL1-4 mRNA transcripts in placenta was significantly increased compared with appropriate for gestational age (AGA) newborns [85]. Women with LGA newborns had an increased BMI before pregnancy (25 kg/m2), an increased gestational weight gain (19 kg), and increased placental weight (777.6 g) compared to AGA newborns associated with a normal maternal BMI before pregnancy of 22.4 kg/m2, a gestational weight gain of 15.8 kg, and a placental weight of 650 g, respectively [85].

mTORC1 promotes placental nutrient transfer

The placenta is the nutrient and endocrine system controlling prenatal mTORC1 signaling for appropriate fetal growth [97,98]. The syncytiotrophoblast, which highly expresses mTOR [99], represents the transporting epithelium and the primary endocrine cell of the human placenta and functions as an mTORC1-dependent nutrient sensor that plays a unique role in the regulation of fetal growth [100]. It has been demonstrated in cultured primary human trophoblast cells that mTORC1 is regulated by glucose, amino acids, and growth factors [101]. mTORC1 is a positive regulator of placental system A and system L amino acid transporters, suggesting that trophoblast mTORC1 modulates amino acid transfer across the placenta [100]. Trophoblast mTORC1 activation increases the cell surface density of amino acid transporters and thus links maternal nutrient availability and growth factor signaling to fetal growth by modulating the mTORC1-mediated flux of amino acids across the placenta, a mechanism that finally results fetal overgrowth (Figure 1) [100].
Activation of placental mTORC1 signaling has been observed in association with maternal obesity [102]. In female Albino Wistar rats, maternal overweight increased placental mTOR and fetal growth [103]. Obesity is associated with elevated circulating levels of BCAAs, free palmitate, hyperinsulinemia, and insulin resistance [66-70,104]. Obviously, the metabolomics of obesity with enhanced nutrient and hormonal signals overstimulate trophoblast mTORC1 activity. In fact, in obese women giving birth to LGA newborns, the activity of placental insulin/IGF-1 and mTORC1 signaling was positively correlated with birth weight [103].
In contrast, mTORC1 in the human placenta is downregulated in restricted fetal growth [99]. Furthermore, in pregnant baboons maternal nutrient restriction downregulated placental mTOR, insulin/IGF-1 signaling and nutrient transporters [105].

Milk intake and maternal insulin resistance

Maternal insulin resistance is a physiologic adaptation of pregnancy that limits maternal glucose uptake to ensure an adequate supply of glucose that is shunted to the growing fetus. Hyperinsulinemia and insulin resistance start to develop in the second half of pregnancy and are induced by the placenta-derived growth hormones, placental growth hormone (PGH) and human placental lactogen (HPL). The somatogenic and lactogenic hormones of the placenta and maternal pituitary gland integrate the metabolic adaptations of pregnancy with the demands of fetal and neonatal development. Dysregulation of placental growth hormones in pathologic conditions of pregnancy adversely affects fetal growth and postnatal metabolic function [106]. In addition to promoting growth of maternal tissue, PGH induces maternal insulin resistance and thereby facilitates the mobilization of maternal nutrients for fetal growth. HPL and prolactin increase maternal food intake by induction of central leptin resistance and promote maternal β-cell expansion and insulin production [106]. Remarkably, milk consumption during pregnancy increased placental weight [87,88], which has been associated with increased maternal serum levels of HPL [89-91]. PGH, which activates the maternal GH receptor (GHR), and HPL, which activates the maternal prolactin receptor (PRLR) both induce signal transducer and activator of transcription 5 (STAT5) [107]. STAT5 promotes the expression of suppressor of cytokine signaling proteins (SOCS) [107]. It is well known that HPL stimulates the Janus-activated-kinase-2 (JAK2)/STAT5 signaling pathway [108-110]. HPL induces SOCS1 and SOCS2 [111]. SOCS1, SOCS3, SOCS6 and SOCS7 are negative regulators of insulin signaling by binding to the insulin receptor (IR), blocking access of signaling intermediates and inhibiting IR tyrosine kinase activity, leading to a reduction of IR-directed phosphorylation of IRS-1 and its downstream events, and by targeting IRS-1 and IRS-2 for proteasomal degradation [112-114]. Increased PGH and HPL signaling via upregulated SOCS expression thus induces SOCS-mediated insulin resistance (Figure 1) [112-115].
Overstimulated mTORC1 signaling activates S6K1 [7,14,116,117], which reduces insulin signaling by inhibitory phosphorylation of IRS-1 [116,117]. BCAA-mediated insulin resistance is explained by enhanced activation of S6K1 [117-124]. In fact, high intake of milk, but not meat, induces insulin resistance in humans [58].
Milk consumption during pregnancy apparently increases the magnitude of maternal insulin resistance 1) by upregulation of placental HPL-SOCS signaling, and 2) by stimulation of maternal mTORC1-S6K1 signaling (Figure 1). Both pathways in a synergistic manner may enhance the magnitude of maternal insulin resistance, thereby increasing the glucose flux to the fetus.

Milk and FGF21-mediated GLUT1-overexpression

Placental weight gain, which is related to milk consumption during pregnancy, is associated with increased maternal serum levels of HPL [87-91]. HPL activates downstream JAK2/STAT5 signaling [108-110]. Recently, fibroblast growth factor-21 (FGF21) has been related to insulin resistance, type 2 diabetes mellitus, obesity and the metabolic syndrome [125,126]. In comparison to control subjects, plasma FGF21 levels were significantly higher in women with gestational diabetes mellitus (GDM) [127]. Increased mRNA expression of FGF21 has been detected in the placenta of women with GDM [128]. Notably, the FGF21 promoter contains three putative STAT5-binding sites [129]. Increased FGF21 production has been observed in late pregnancy in the mouse [130]. Ectopic activation of hepatic mTORC1 in liver-specific Tsc1 knockout mice resulted in enhanced expression of FGF21 [131]. Intriguingly, overexpression of FGF21 in 3 T3-L1 adipocytes upregulated glucose uptake and increased mRNA expression of glucose transporter 1 (GLUT1) [132]. GLUT1 is the primary glucose transporter isoform in the human placenta that increases its expression over gestation [133]. GLUT1 has been localized to both the maternal facing microvillous plasma membrane (MVM) with threefold higher expression as compared to the basal plasma membrane (BM) [134]. In maternal diabetes, the expression of GLUT1 in the BM has been reported to increase [135,136]. Moreover, increased BM expression of GLUT1 has been associated with high birth weight of large babies of non-diabetic mothers [137].
Thus, milk-mediated overactivation of mTORC1 via placental overexpression of FGF21 and enhanced HPL/STAT5-driven placental expression of FGF21 may overstimulate trophoblast GLUT1 expression that increases the diaplacental flux of glucose to the fetus (Figure 1). Alterations of maternal and placental metabolic signaling by milk consumption during pregnancy may thus explain accelerated fetal growth and increased birth weight.

MicroRNA-21 and placental, fetal and adipocyte growth

Jiang et al. [138] recently reported on aberrant upregulation of microRNA-21 in placental tissue of macrosomia. Importantly, exosomal microRNA-21 is an abundant and consistent microRNA of cow’s milk [139]. Notably, human and bovine microRNA-21 stem-loops are identical (www.​mirbase.​org). Milk has been proposed to function as a metabolic transfection system by transfer of exosomal microRNAs activating mTORC1 signaling of the milk recipient [2]. Milk’s exosomal microRNA represent milk’s “software” and milk-derived BCAAs milk’s “hardware” for activating mTORC1 signaling [2]. In fact, Baier et al. [140] provided evidence that microRNAs of commercial pasteurized cow’s milk are absorbed by adult human subjects in biologically meaningful amounts from nutritionally relevant doses of cow’s milk and affect gene expression of peripheral blood mononuclear cells, HEK-293 kidney cell cultures and mouse livers. Furthermore, they demonstrated that disintegration of milk exosomes by ultrasonication abolished the transfer and uptake of milk-derived microRNAs [140]. Notably, in placental tissues target genes of microRNA-21 were involved in JAK-STAT, PI3K-AKT, and mTOR signaling pathways [138]. It is thus conceivable that milk-derived exosomal microRNA-21 may reach the trophoblast cell and contributes to overactivated trophoblast mTORC1 signaling. Critical targets of microRNA-21 are mRNAs of important tumor suppressor proteins involved in upstream and downstream suppression of mTORC1 signaling such as PTEN [141-144], Sprouty1 and Sprouty2 [145-147], and PDCD4 [148-150]. Moreover, microRNA-21 has been shown to induce the cell cycle promoter cyclin D1 in an mTORC1-dependent manner [151]. Supposed that milk-derived microRNA-21 reaches the trophoblast cells via systemic circulation of the pregnant milk-consuming mother, PTEN suppression could increase insulin/IGF-1/PI3K/AKT signaling, which further augments mTORC1 activation (Figure 1). MicroRNA-21-mediated inhibition of Sprouty1 and 2 would amplify RAS-RAF-MEK-ERK signaling, which additionally suppresses TSC2 and thus raises mTORC1 activity. Furthermore, microRNA-21 could stimulate the initiation of translation by repression of PDCD4, which is a suppressor of translation initiation that inhibits the RNA helicase eIF4A [152]. Both, 4E-BP-1 and PDCD4 are crucial regulatory inhibitors of translation initiation and thus of protein synthesis. Activation of the mTORC1 pathway and its substrate kinase S6K1 results in subsequent phosphorylation of 4E-BP-1 and PDCD4 that promote eIF4E-eIF4G complex assembly and stimulate mRNA translation [151]. Milk microRNA-21-mediated suppression of PDCD4 expression may further amply translation, which enhances placental and finally fetal overgrowth. Furthermore, microRNA-21 promotes adipogenic differentiation and proliferation of human adipose tissue-derived mesenchymal stem cells [153,154], thus promoting fat mass accretion. It has recently been shown that long-term inhibition of microRNA-21 reduced obesity in db/db mice [155].

Milk consumption and fetal and birth weight

The Generation R Study, a population-based prospective cohort study from fetal life until young adulthood in Rotterdam investigated 3 405 mothers during pregnancy [156]. Maternal milk consumption of >3 glasses (450 mL of milk) per day was associated with greater fetal weight gain in the third trimester of pregnancy, which led to an 88 g higher birth weight than that with milk consumption of 0 to 1 glass per day [156]. This association was limited to milk, whereas protein intake from nondairy food or cheese was not associated with increased birth weight. A possible explanation for this finding is the presence of biologically active microRNAs in milk and their absence in processed milk products such as cheese. Compared with the lowest reference category of milk consumption (0–1 glasses/day), maternal milk intakes of > 1–2 glasses/day, 2–3 glasses/day, and > 3 glasses/day were associated with increased fetal weight gain. Fetal weight gain has been estimated by the procedure of Hadlock et al. [157]. Milk-mediated differences in fetal weight gain appeared from the 20th week onward, but became most evident in the last part of the third trimester [156], a period that is controlled by HLP signaling.
Worldwide studies confirmed an increase of birth weight in relation to milk consumption during pregnancy (Table 1). A retrospective cohort in Sweden reported a birth weight increase of 75 g and 134 g in the offspring of mothers consuming > 200 mL and 1 L milk daily, respectively [158]. A prospective study in India reported that the frequency of milk consumption at the 18th week of gestation was positively associated with birth weight, birth length, and head circumference [88]. According to a prospective study in Canada, maternal daily consumption of an additional 237 mL of milk was associated with a 41 g increase in offspring birth weight [159]. A prospective Australian study in 557 mothers reported that protein intake from dairy products was associated with a higher birth weight of the offspring [160]. In a randomized controlled trial of 72 adolescent pregnant mothers in the USA, 25 mothers were counseled to consume > 4 servings of dairy products a day, which resulted in a 240 g higher birth weight compared to the control group [161]. According to a systematic literature review, the majority of studies reported positive associations between milk and/or dairy consumption and birth weight-related outcomes [162].
Table 1
Milk intake increases prepregnancy, gestational, placental, fetal, and birth weight
Effect of milk intake
Outcome
Studies [Ref.]
Prepregnancy weight
Increase
Randomized intervention study, Denmark [80]
Gestational weight gain
Increase
Observational study, Iceland [86]
Placental weight
Increase
Danish National Birth Cohort, Denmark [87]
Increase
Pune Maternal Nutrition Study, India [88]
Fetal weight
Increase
Generation R Study, Netherlands [156]
Birth weight
Increase
Generation R Study, Netherlands [156]
Increase
Observational study, Sweden [158]
Increase
Pune Maternal Nutrition Study, India [88]
Increase
Observational study, Canada [159]
Increase
Prospective observational study, Australia [160]
Increase
Randomized controlled trial, USA [161]
Increase
Systematic literature review, Norway [162]

Conclusions

We provided literature evidence supported by translational research that milk consumption increases pregravid, gestational, placental, fetal, and birth weight, respectively (Table 1). The Mater-University Study of Pregnancy and Its Outcomes demonstrated that maternal BMI in comparison to paternal BMI is associated with the BMI of the offspring at ages 5 and 14 years [22]. Based on these data, Lawlor et al. [22] proposed the fetal overnutrition hypothesis of obesity and concluded that nutrient-dependent programming effects during fetal life are responsible for the development of obesity [22].
The central hub of nutrient sensing, growth regulation and anabolism is the kinase mTORC1, which is upregulated in obese subjects and by milk consumption [2,14-17]. Milk is an evolutionary feeding and anabolic programming system controlled by the lactation genome that regulates mTORC1-dependent postnatal growth by donation of mTORC1-activating essential BCAAs and exosomal microRNAs [2,140]. The placenta is the natural feeding and programming system controlling mTORC1-dependent fetal growth. No other gravid mammal is simultaneously exposed to lactation-driven as well as placenta-driven mTORC1 signaling, except human beings since the Neolithic revolution boosted after the widespread distribution of refrigerators in the early 1950’s allowing daily access to bovine milk. From an anthropological perspective, Wiley [13] concluded that milk consumption by humans is a novel behavior that increases BMI and may induce long-lasting adverse effects on human health. In fact, our evidence underlines that milk consumption increases prepregnancy BMI [79-81], gestational [85,86], placental [87,88], fetal [156], and birth weight [156,158-162], respectively. Notably, increased birth weight, is a risk factor for the development of mTORC1-driven diseases of civilization [163-171]. The magnitude of fetal and postnatal mTORC1-signaling apparently determines lifelong axes of metabolic, hypothalamic and immunological programming [172-176].
Intrauterine overnutrition affects the risk of obesity [177-180]. High maternal plasma concentrations of glucose, amino acids and free fatty acids have been implicated to result in permanent changes in appetite control, neuroendocrine functioning, and energy metabolism in the developing fetus, thus leading to obesity later in life [176-179]. Milk consumption provides abundant BCAAs and palmitate, stimulates insulin/IGF-1 signaling, and provides abundant exosomal microRNAs that in a synergistic manner may overstimulate trophoblast mTORC1 activity (Figure 1). Overactivated trophoblast mTORC1 signaling finally explains 1) increased expression of mTORC1-dependent amino acid transporters with enhanced diaplacental flux of amino acids, 2) increased HPL synthesis with STAT5-promoted induction of maternal insulin resistance thus increasing the glucose flux to the fetus, and 3) increased STAT5/FGF21- and mTORC1/FGF21-driven trophoblast GLUT1 expression promoting diaplacental glucose transfer to the fetus. Accordingly, fetal cells obtain an excessive supply of glucose and BCAAs. Finally, BCAAs that reach fetal cells overactivate fetal mTORC1 signaling promoting fetal overgrowth [180].
When overactivated mTORC1 signaling persists during the postnatal period by the introduction of artificial high protein formula feeding, lifelong deviations of mTORC1-dependent metabolic, neuroendocrine and immunological programming may result [181,182]. In this regard, the worst scenario for mTORC1-dependent perinatal malprogramming is an obese mother, who increases milk consumption during pregnancy, and provides excessive protein by artificial formula feeding [182]. High milk intake during pregnancy and high protein formula feeding may synergistically enhance perinatal mTORC1 signaling explaining the fetal overnutrition hypothesis and the early protein hypothesis [22,174,182]. These two hypotheses converge to a perinatal mTORC1-overactivation hypothesis, explaining the adverse effects of increased milk-mediated mTORC1 signaling during the pre- and postnatal period of metabolic programming.
Current dietary recommendations for pregnant women intend to assure sufficient supply of calcium and high quality proteins for the growing fetus. However, there is more and more concern about milk’s role as a source of calcium. According to the recent opinion of Harvard School of Public Health milk isn’t the only, or even best, source of calcium [183]. There are non-dairy foods including leafy green vegetables, broccoli, beans and tofu that supply high amounts of calcium. These calcium-rich food alternatives have a significant advantage in comparison to milk: they do not overstimulate mTORC1 signaling and most importantly do not transfer biologically active exosomal microRNAs [141].
Therefore, we suggest to re-evaluate dietary recommendations for pregnant women. We appeal to the medical community to define save upper limits for milk consumption during pregnancy, especially for those women who enter gravity with increased BMI. Whereas boiling of milk destroys milk’s bioactive microRNAs [184], boiling has no effect on milk-BCAA-mediated mTORC1 activation. Future randomized-controlled clinical studies are needed to better study the effect of dietary interventions based on milk consumption’s difference during pregnancy, especially in women who enter pregnancy with overweight or obesity, and the risk of increased birth weight [185].

Acknowledgements

We would like to thank Gerda Sternecker and Daniela Wagenknecht for the acquisition of literature.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

BCM was responsible for translational research and data abstraction. All authors performed and contributed to literature search, reviewed and edited the manuscript. All authors read and approved the final manuscript.
Literatur
2.
Zurück zum Zitat Melnik BC, John SM, Schmitz G. Milk is not just food but most likely a genetic transfection system activating mTORC1 for postnatal growth. Nutr J. 2013;12:103.PubMedCentralPubMed Melnik BC, John SM, Schmitz G. Milk is not just food but most likely a genetic transfection system activating mTORC1 for postnatal growth. Nutr J. 2013;12:103.PubMedCentralPubMed
3.
Zurück zum Zitat Foster KG, Fingar DC. Mammalian target of rapamycin (mTOR): conducting the cellular signaling symphony. J Biol Chem. 2010;285:14071–7.PubMedCentralPubMed Foster KG, Fingar DC. Mammalian target of rapamycin (mTOR): conducting the cellular signaling symphony. J Biol Chem. 2010;285:14071–7.PubMedCentralPubMed
4.
Zurück zum Zitat Inoki K, Ouyang H, Li Y, Guan KL. Signaling by target of rapamycin proteins in cell growth control. Microbiol Mol Biol Rev. 2005;69:79–100.PubMedCentralPubMed Inoki K, Ouyang H, Li Y, Guan KL. Signaling by target of rapamycin proteins in cell growth control. Microbiol Mol Biol Rev. 2005;69:79–100.PubMedCentralPubMed
5.
Zurück zum Zitat Avruch J, Long X, Ortiz-Vega S, Rapley J, Papageorgiou A, Dai N. Amino acid regulation of TOR complex 1. Am J Physiol Endocrinol Metab. 2009;296:E592–602.PubMedCentralPubMed Avruch J, Long X, Ortiz-Vega S, Rapley J, Papageorgiou A, Dai N. Amino acid regulation of TOR complex 1. Am J Physiol Endocrinol Metab. 2009;296:E592–602.PubMedCentralPubMed
6.
Zurück zum Zitat Sengupta S, Peterson T, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell. 2010;40:310–22.PubMedCentralPubMed Sengupta S, Peterson T, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell. 2010;40:310–22.PubMedCentralPubMed
7.
Zurück zum Zitat Laplante M, Sabatini DM. mTOR signaling. Cold Spring Harb Perspect Biol. 2012; 4. Laplante M, Sabatini DM. mTOR signaling. Cold Spring Harb Perspect Biol. 2012; 4.
8.
Zurück zum Zitat Kim J, Guan KL. Amino acid signaling in TOR activation. Annu Rev Plant Physiol Plant Mol Biol. 2011;80:1001–32. Kim J, Guan KL. Amino acid signaling in TOR activation. Annu Rev Plant Physiol Plant Mol Biol. 2011;80:1001–32.
9.
Zurück zum Zitat Kim S, Buel GR, Blenis J. Nutrient regulation of the mTOR complex 1 signaling pathway. Mol Cells. 2013;35:463–73.PubMedCentralPubMed Kim S, Buel GR, Blenis J. Nutrient regulation of the mTOR complex 1 signaling pathway. Mol Cells. 2013;35:463–73.PubMedCentralPubMed
10.
11.
Zurück zum Zitat Efeyan A, Sabatini DM. Nutrients and growth factors in mTORC1 activation. Biochem Soc Trans. 2013;41:902–5.PubMed Efeyan A, Sabatini DM. Nutrients and growth factors in mTORC1 activation. Biochem Soc Trans. 2013;41:902–5.PubMed
12.
Zurück zum Zitat Laplante M, Sabatini DM. Regulation of mTORC1 and its impact on gene expression at a glance. J Cell Sci. 2013;126:1713–9.PubMedCentralPubMed Laplante M, Sabatini DM. Regulation of mTORC1 and its impact on gene expression at a glance. J Cell Sci. 2013;126:1713–9.PubMedCentralPubMed
13.
Zurück zum Zitat Wiley AS. Cow milk consumption, insulin-like growth factor-I, and human biology: a life history approach. Am J Hum Biol. 2012;24:130–8.PubMed Wiley AS. Cow milk consumption, insulin-like growth factor-I, and human biology: a life history approach. Am J Hum Biol. 2012;24:130–8.PubMed
14.
Zurück zum Zitat Yamin HB, Barnea M, Genzer Y, Chapnik N, Froy O. Long-term commercial cow’s milk consumption and its effects on metabolic parameters associated with obesity in young mice. Mol Nutr Food Res. 2014;58:1061–8.PubMed Yamin HB, Barnea M, Genzer Y, Chapnik N, Froy O. Long-term commercial cow’s milk consumption and its effects on metabolic parameters associated with obesity in young mice. Mol Nutr Food Res. 2014;58:1061–8.PubMed
15.
Zurück zum Zitat Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nature Rev Mol Cell Biol. 2011;12:21–35. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nature Rev Mol Cell Biol. 2011;12:21–35.
16.
Zurück zum Zitat Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013;493:338–45.PubMedCentralPubMed Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013;493:338–45.PubMedCentralPubMed
17.
18.
Zurück zum Zitat Symonds ME, Mendez MA, Meltzer HM, Koletzko B, Godfrey K, Forsyth S, et al. Early life nutritional programming of obesity: mother-child cohort studies. Ann Nutr Metab. 2013;62:137–45.PubMed Symonds ME, Mendez MA, Meltzer HM, Koletzko B, Godfrey K, Forsyth S, et al. Early life nutritional programming of obesity: mother-child cohort studies. Ann Nutr Metab. 2013;62:137–45.PubMed
19.
Zurück zum Zitat Abrams BF, Laros Jr RK. Prepregnancy weight, weight gain, and birth weight. Am J Obstet Gynecol. 1986;154:503–9.PubMed Abrams BF, Laros Jr RK. Prepregnancy weight, weight gain, and birth weight. Am J Obstet Gynecol. 1986;154:503–9.PubMed
20.
Zurück zum Zitat O’Callaghan MJ, Williams GM, Andersen MJ, Bor W, Najman JM. Prediction of obesity in children at 5 years: a cohort study. J Paediatr Child Health. 1997;33:311–6.PubMed O’Callaghan MJ, Williams GM, Andersen MJ, Bor W, Najman JM. Prediction of obesity in children at 5 years: a cohort study. J Paediatr Child Health. 1997;33:311–6.PubMed
21.
Zurück zum Zitat Li C, Kaur H, Choi WS, Huang TT, Lee RE, Ahluwalia JS. Additive interactions of maternal prepregnancy BMI and breast-feeding on childhood overweight. Obes Res. 2005;13:362–71.PubMed Li C, Kaur H, Choi WS, Huang TT, Lee RE, Ahluwalia JS. Additive interactions of maternal prepregnancy BMI and breast-feeding on childhood overweight. Obes Res. 2005;13:362–71.PubMed
22.
Zurück zum Zitat Lawlor DA, Smith GD, O’Callaghan M, Alati R, Mamun AA, Williams GM, et al. Epidemiologic evidence for the fetal overnutrition hypothesis: findings from the Mater-University Study of Pregnancy and its Outcomes. Am J Epidemiol. 2007;165:418–24.PubMed Lawlor DA, Smith GD, O’Callaghan M, Alati R, Mamun AA, Williams GM, et al. Epidemiologic evidence for the fetal overnutrition hypothesis: findings from the Mater-University Study of Pregnancy and its Outcomes. Am J Epidemiol. 2007;165:418–24.PubMed
23.
Zurück zum Zitat Davey Smith G, Steer C, Leary S, Ness A. Is there an intrauterine influence on obesity? Evidence from parent child associations in the Avon Longitudinal Study of Parents and Children (ALSPAC). Arch Dis Child. 2007;92:876–80.PubMed Davey Smith G, Steer C, Leary S, Ness A. Is there an intrauterine influence on obesity? Evidence from parent child associations in the Avon Longitudinal Study of Parents and Children (ALSPAC). Arch Dis Child. 2007;92:876–80.PubMed
24.
Zurück zum Zitat Viswanathan M, Siega-Riz AM, Moos MK, Deierlein A, Mumford S, Knaack J, et al. Outcomes of maternal weight gain. Evid Rep Technol Assess (Full Rep). 2008;168:1–223. Viswanathan M, Siega-Riz AM, Moos MK, Deierlein A, Mumford S, Knaack J, et al. Outcomes of maternal weight gain. Evid Rep Technol Assess (Full Rep). 2008;168:1–223.
25.
Zurück zum Zitat Catalano PM, Farrell K, Thomas A, Huston-Presley L, Mencin P, de Mouzon SH, et al. Perinatal risk factors for childhood obesity and metabolic dysregulation. Am J Clin Nutr. 2009;90:1303–13.PubMedCentralPubMed Catalano PM, Farrell K, Thomas A, Huston-Presley L, Mencin P, de Mouzon SH, et al. Perinatal risk factors for childhood obesity and metabolic dysregulation. Am J Clin Nutr. 2009;90:1303–13.PubMedCentralPubMed
26.
Zurück zum Zitat Jääskeläinen A, Pussinen J, Nuutinen O, Schwab U, Pirkola J, Kolehmainen M, et al. Intergenerational transmission of overweight among Finnish adolescents and their parents: a 16-year follow-up study. Int J Obes (Lond). 2011;35:1289–94. Jääskeläinen A, Pussinen J, Nuutinen O, Schwab U, Pirkola J, Kolehmainen M, et al. Intergenerational transmission of overweight among Finnish adolescents and their parents: a 16-year follow-up study. Int J Obes (Lond). 2011;35:1289–94.
27.
Zurück zum Zitat Wang X, Proud CG. Nutrient control of mTORC1, a cell-cycle regulator. Trends Cell Biol. 2009;19:260–7.PubMed Wang X, Proud CG. Nutrient control of mTORC1, a cell-cycle regulator. Trends Cell Biol. 2009;19:260–7.PubMed
28.
Zurück zum Zitat Chakrabarti P, English T, Shi J, Smas CM, Kandror KV. Mammalian target of rapamycin complex 1 suppresses lipolysis, stimulates lipogenesis, and promotes fat storage. Diabetes. 2010;59:775–81.PubMedCentralPubMed Chakrabarti P, English T, Shi J, Smas CM, Kandror KV. Mammalian target of rapamycin complex 1 suppresses lipolysis, stimulates lipogenesis, and promotes fat storage. Diabetes. 2010;59:775–81.PubMedCentralPubMed
29.
Zurück zum Zitat Ricoult SJ, Manning BD. The multifaceted role of mTORC1 in the control of lipid metabolism. EMBO Rep. 2013;14:242–51.PubMedCentralPubMed Ricoult SJ, Manning BD. The multifaceted role of mTORC1 in the control of lipid metabolism. EMBO Rep. 2013;14:242–51.PubMedCentralPubMed
30.
Zurück zum Zitat Howell JJ, Ricoult SJ, Ben-Sahra I, Manning BD. A growing role for mTOR in promoting anabolic metabolism. Biochem Soc Trans. 2013;41:906–12.PubMed Howell JJ, Ricoult SJ, Ben-Sahra I, Manning BD. A growing role for mTOR in promoting anabolic metabolism. Biochem Soc Trans. 2013;41:906–12.PubMed
31.
Zurück zum Zitat Xu J, Ji J, Yan XH. Cross-talk between AMPK and mTOR in regulating energy balance. Crit Rev Food Sci Nutr. 2012;52:373–81.PubMed Xu J, Ji J, Yan XH. Cross-talk between AMPK and mTOR in regulating energy balance. Crit Rev Food Sci Nutr. 2012;52:373–81.PubMed
32.
Zurück zum Zitat Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A. 2004;101:3329–35.PubMedCentralPubMed Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A. 2004;101:3329–35.PubMedCentralPubMed
33.
Zurück zum Zitat Dodd KM, Tee AR. Leucine and mTORC1: a complex relationship. Am J Physiol Endocrinol Metab. 2012;302:E1329–42.PubMed Dodd KM, Tee AR. Leucine and mTORC1: a complex relationship. Am J Physiol Endocrinol Metab. 2012;302:E1329–42.PubMed
34.
Zurück zum Zitat Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell. 2009;136:521–34.PubMedCentralPubMed Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell. 2009;136:521–34.PubMedCentralPubMed
35.
Zurück zum Zitat Cohen A, Hall MN. An amino acid shuffle activates mTORC1. Cell. 2009;136:399–400.PubMed Cohen A, Hall MN. An amino acid shuffle activates mTORC1. Cell. 2009;136:399–400.PubMed
36.
Zurück zum Zitat Duran RV, Oppliger W, Robitaille AM, Heiserich L, Skendaj R, Gottlieb E, et al. Glutaminolysis activates Rag-mTORC1 signaling. Mol Cell. 2012;47:349–58.PubMed Duran RV, Oppliger W, Robitaille AM, Heiserich L, Skendaj R, Gottlieb E, et al. Glutaminolysis activates Rag-mTORC1 signaling. Mol Cell. 2012;47:349–58.PubMed
37.
Zurück zum Zitat Yasuda M, Tanaka Y, Kume S, Morita Y, Chin-Kanasaki M, Araki H, et al. Fatty acids are novel nutrient factors to regulate mTORC1 lysosomal localization and apoptosis in podocytes. Biochim Biophys Acta. 2014;1842:1097–108.PubMed Yasuda M, Tanaka Y, Kume S, Morita Y, Chin-Kanasaki M, Araki H, et al. Fatty acids are novel nutrient factors to regulate mTORC1 lysosomal localization and apoptosis in podocytes. Biochim Biophys Acta. 2014;1842:1097–108.PubMed
38.
Zurück zum Zitat Millward DJ, Layman DK, Tomé D, Schaafsma G. Protein quality assessment: impact of expanding understanding of protein and amino acid needs for optimal health. Am J Clin Nutr. 2008;87:1576S–81S.PubMed Millward DJ, Layman DK, Tomé D, Schaafsma G. Protein quality assessment: impact of expanding understanding of protein and amino acid needs for optimal health. Am J Clin Nutr. 2008;87:1576S–81S.PubMed
39.
Zurück zum Zitat Boirie Y, Dangin M, Gachon P, Vasson MP, Maubois JL, Beaufrère B. Slow and fast dietary proteins differently modulate postprandial protein accretion. Proc Natl Acad Sci U S A. 1997;94:14930–5.PubMedCentralPubMed Boirie Y, Dangin M, Gachon P, Vasson MP, Maubois JL, Beaufrère B. Slow and fast dietary proteins differently modulate postprandial protein accretion. Proc Natl Acad Sci U S A. 1997;94:14930–5.PubMedCentralPubMed
40.
Zurück zum Zitat He T, Giuseppin ML. Slow and fast dietary proteins differentially modulate postprandial metabolism. Int J Food Sci Nutr. 2014;65:386–90.PubMed He T, Giuseppin ML. Slow and fast dietary proteins differentially modulate postprandial metabolism. Int J Food Sci Nutr. 2014;65:386–90.PubMed
41.
Zurück zum Zitat Boutrou R, Gaudichon C, Dupont D, Jardin J, Airinei G, Marsset-Baglieri A, et al. Sequential releases of milk protein-derived bioactive peptides in the jejunum in healthy humans. Am J Clin Nutr. 2013;97:1414–23. Boutrou R, Gaudichon C, Dupont D, Jardin J, Airinei G, Marsset-Baglieri A, et al. Sequential releases of milk protein-derived bioactive peptides in the jejunum in healthy humans. Am J Clin Nutr. 2013;97:1414–23.
42.
Zurück zum Zitat Mahé S, Roos N, Benamouzig R, Davin L, Luengo C, Gagnon L, et al. Gastrojejunal kinetics and the digestion of [15 N]beta-lactoglobulin and casein in humans: the influence of the nature and quantity of the protein. Am J Clin Nutr. 1996;63:546–52.PubMed Mahé S, Roos N, Benamouzig R, Davin L, Luengo C, Gagnon L, et al. Gastrojejunal kinetics and the digestion of [15 N]beta-lactoglobulin and casein in humans: the influence of the nature and quantity of the protein. Am J Clin Nutr. 1996;63:546–52.PubMed
43.
Zurück zum Zitat Lenders CM, Liu S, Wilmore DW, Sampson L, Dougherty LW, Spiegelman D, et al. Evaluation of a novel food composition database that includes glutamine and other amino acids derived from gene sequencing data. Eur J Clin Nutr. 2009;63:1433–9.PubMedCentralPubMed Lenders CM, Liu S, Wilmore DW, Sampson L, Dougherty LW, Spiegelman D, et al. Evaluation of a novel food composition database that includes glutamine and other amino acids derived from gene sequencing data. Eur J Clin Nutr. 2009;63:1433–9.PubMedCentralPubMed
44.
Zurück zum Zitat Li M, Li C, Allen A, Stanley CA, Smith TJ. The structure and allosteric regulation of mammalian glutamate dehydrogenase. Arch Biochem Biophys. 2012;519:69–80.PubMedCentralPubMed Li M, Li C, Allen A, Stanley CA, Smith TJ. The structure and allosteric regulation of mammalian glutamate dehydrogenase. Arch Biochem Biophys. 2012;519:69–80.PubMedCentralPubMed
45.
Zurück zum Zitat Lorin S, Tol MJ, Bauvy C, Strijland A, Poüs C, Verhoeven AJ, et al. Glutamate dehydrogenase contributes to leucine sensing in the regulation of autophagy. Autophagy. 2013;9:850–60.PubMedCentralPubMed Lorin S, Tol MJ, Bauvy C, Strijland A, Poüs C, Verhoeven AJ, et al. Glutamate dehydrogenase contributes to leucine sensing in the regulation of autophagy. Autophagy. 2013;9:850–60.PubMedCentralPubMed
46.
Zurück zum Zitat Xu G, Kwon G, Cruz WS, Marshall CA, McDaniel ML. Metabolic regulation by leucine of translation initiation through the mTOR-signaling pathway by pancreatic beta-cells. Diabetes. 2001;50:353–60.PubMed Xu G, Kwon G, Cruz WS, Marshall CA, McDaniel ML. Metabolic regulation by leucine of translation initiation through the mTOR-signaling pathway by pancreatic beta-cells. Diabetes. 2001;50:353–60.PubMed
47.
Zurück zum Zitat Holt S, Brand Miller J, Petocz P. An insulin index of foods: the insulin demand generated by 1000-kK portions of common foods. Am J Clin Nutr. 1997;66:1264–76.PubMed Holt S, Brand Miller J, Petocz P. An insulin index of foods: the insulin demand generated by 1000-kK portions of common foods. Am J Clin Nutr. 1997;66:1264–76.PubMed
48.
Zurück zum Zitat Hoyt G, Hickey MS, Cordain L. Dissociation of the glycaemic and insulinaemic responses to whole and skimmed milk. Br J Nutr. 2005;93:175–7.PubMed Hoyt G, Hickey MS, Cordain L. Dissociation of the glycaemic and insulinaemic responses to whole and skimmed milk. Br J Nutr. 2005;93:175–7.PubMed
49.
Zurück zum Zitat Hoppe C, Mølgaard C, Dalum C, Vaag A, Michaelsen KF. Differential effects of casein versus whey on fasting plasma levels of insulin, IGF-1 and IGF-1/IGFBP-3: results from a randomized 7-day supplementation study in prepubertal boys. Eur J Clin Nutr. 2009;63:1076–83.PubMed Hoppe C, Mølgaard C, Dalum C, Vaag A, Michaelsen KF. Differential effects of casein versus whey on fasting plasma levels of insulin, IGF-1 and IGF-1/IGFBP-3: results from a randomized 7-day supplementation study in prepubertal boys. Eur J Clin Nutr. 2009;63:1076–83.PubMed
50.
Zurück zum Zitat Thomas FB, Sinar D, Mazzaferri EL, Cataland S, Mekhjian HS, Caldwell JH, et al. Selective release of gastric inhibitory polypeptide by intraduodenal amino acid perfusion in man. Gastroenterology. 1978;74:1261–5.PubMed Thomas FB, Sinar D, Mazzaferri EL, Cataland S, Mekhjian HS, Caldwell JH, et al. Selective release of gastric inhibitory polypeptide by intraduodenal amino acid perfusion in man. Gastroenterology. 1978;74:1261–5.PubMed
51.
Zurück zum Zitat Chen Q, Reimer RA. Dairy protein and leucine alter GLP-1 release and mRNA of genes involved in intestinal lipid metabolism in vitro. Nutrition. 2009;25:340–9.PubMed Chen Q, Reimer RA. Dairy protein and leucine alter GLP-1 release and mRNA of genes involved in intestinal lipid metabolism in vitro. Nutrition. 2009;25:340–9.PubMed
52.
Zurück zum Zitat Nilsson M, Stenberg M, Frid AH, Holst JJ, Björck IM. Glycemia and insulinemia in healthy subjects after lactose-equivalent meals of milk and other food proteins: the role of plasma amino acids and incretins. Am J Clin Nutr. 2004;80:1246–53.PubMed Nilsson M, Stenberg M, Frid AH, Holst JJ, Björck IM. Glycemia and insulinemia in healthy subjects after lactose-equivalent meals of milk and other food proteins: the role of plasma amino acids and incretins. Am J Clin Nutr. 2004;80:1246–53.PubMed
53.
Zurück zum Zitat Nilsson M, Holst JJ, Björck IM. Metabolic effects of amino acid mixtures and whey protein in helathy subjects: studies using glucose-equivalent drinks. Am J Clin Nutr. 2007;85:996–1004.PubMed Nilsson M, Holst JJ, Björck IM. Metabolic effects of amino acid mixtures and whey protein in helathy subjects: studies using glucose-equivalent drinks. Am J Clin Nutr. 2007;85:996–1004.PubMed
54.
Zurück zum Zitat Salehi A, Gunnerud U, Muhammed SJ, Ostman E, Holst JJ, Björck I, et al. The insulinogenic effects of whey protein is partially mediated by a direct effect of amino acids and GIP on β-cells. Nutr Metab (Lond). 2012;9:48. Salehi A, Gunnerud U, Muhammed SJ, Ostman E, Holst JJ, Björck I, et al. The insulinogenic effects of whey protein is partially mediated by a direct effect of amino acids and GIP on β-cells. Nutr Metab (Lond). 2012;9:48.
55.
Zurück zum Zitat McDaniel ML, Marshall CA, Pappan KL, Kwon G. Metabolic and autocrine regulation of the mammalian target of rapamycin by pancreatic β-cells. Diabetes. 2002;51:2877–85.PubMed McDaniel ML, Marshall CA, Pappan KL, Kwon G. Metabolic and autocrine regulation of the mammalian target of rapamycin by pancreatic β-cells. Diabetes. 2002;51:2877–85.PubMed
56.
Zurück zum Zitat Yang J, Chi Y, Burkhardt BR, Guan Y, Wolf BA. Leucine metabolism in regulation of insulin secretion from pancreatic beta cells. Nutr Rev. 2010;68:270–9.PubMedCentralPubMed Yang J, Chi Y, Burkhardt BR, Guan Y, Wolf BA. Leucine metabolism in regulation of insulin secretion from pancreatic beta cells. Nutr Rev. 2010;68:270–9.PubMedCentralPubMed
57.
Zurück zum Zitat Le Bacquer O, Queniat G, Gmyr V, Kerr-Conte J, Lefebvre B, Pattou F. mTORC1 and mTORC2 regulate insulin secretion through Akt in INS-1 cells. J Endocrinol. 2013;216:21–9.PubMed Le Bacquer O, Queniat G, Gmyr V, Kerr-Conte J, Lefebvre B, Pattou F. mTORC1 and mTORC2 regulate insulin secretion through Akt in INS-1 cells. J Endocrinol. 2013;216:21–9.PubMed
58.
Zurück zum Zitat Hoppe C, Mølgaard C, Vaag A, Barkholt V, Michaelsen KF. High intakes of milk, but not meat, increases s-insulin and insulin resistance in 8-year-old boys. Eur J Clin Nutr. 2005;59:393–8.PubMed Hoppe C, Mølgaard C, Vaag A, Barkholt V, Michaelsen KF. High intakes of milk, but not meat, increases s-insulin and insulin resistance in 8-year-old boys. Eur J Clin Nutr. 2005;59:393–8.PubMed
59.
Zurück zum Zitat Qin LQ, He K, Xu JY. Milk consumption and circulating insulin-like growth factor-I level: a systematic literature review. Int J Food Sci Nutr. 2009;60 Suppl 7:330–40.PubMed Qin LQ, He K, Xu JY. Milk consumption and circulating insulin-like growth factor-I level: a systematic literature review. Int J Food Sci Nutr. 2009;60 Suppl 7:330–40.PubMed
60.
Zurück zum Zitat Norat T, Dossus L, Rinaldi S, Overvad K, Grønbaek H, Tjønneland A, et al. Diet, serum insulin-like growth factor-I and IGF-binding protein-3 in European women. Eur J Clin Nutr. 2007;61:91–8.PubMed Norat T, Dossus L, Rinaldi S, Overvad K, Grønbaek H, Tjønneland A, et al. Diet, serum insulin-like growth factor-I and IGF-binding protein-3 in European women. Eur J Clin Nutr. 2007;61:91–8.PubMed
61.
Zurück zum Zitat Rich-Edwards JW, Ganmaa D, Pollak MN, Nakamoto EK, Kleinman K, Tserendolgor U, et al. Milk consumption and the prepubertal somatotropic axis. Nutr J. 2007;6:28.PubMedCentralPubMed Rich-Edwards JW, Ganmaa D, Pollak MN, Nakamoto EK, Kleinman K, Tserendolgor U, et al. Milk consumption and the prepubertal somatotropic axis. Nutr J. 2007;6:28.PubMedCentralPubMed
62.
Zurück zum Zitat Larnkjær A, Arnberg K, Michaelsen KF, Jensen SM, Mølgaard C. Effect of milk proteins on linear growth and IGF variables in overweight adolescents. Growth Horm IGF Res. 2014;24:54–9.PubMed Larnkjær A, Arnberg K, Michaelsen KF, Jensen SM, Mølgaard C. Effect of milk proteins on linear growth and IGF variables in overweight adolescents. Growth Horm IGF Res. 2014;24:54–9.PubMed
63.
Zurück zum Zitat Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J Diabetes. 2012;3:38–53.PubMedCentralPubMed Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J Diabetes. 2012;3:38–53.PubMedCentralPubMed
64.
Zurück zum Zitat Jensen RG, Ferris AM, Lammi-Keefe CJ. The composition of milk fat. J Dairy Sci. 1991;74:3228–43.PubMed Jensen RG, Ferris AM, Lammi-Keefe CJ. The composition of milk fat. J Dairy Sci. 1991;74:3228–43.PubMed
65.
Zurück zum Zitat Bitman J, Wood DL. Changes in milk fat phospholipids during lactation. J Dairy Sci. 1990;73:1208–16.PubMed Bitman J, Wood DL. Changes in milk fat phospholipids during lactation. J Dairy Sci. 1990;73:1208–16.PubMed
66.
Zurück zum Zitat She P, Van Horn C, Reid T, Hutson SM, Cooney RN, Lynch CJ. Obesity-related elevations in plasma leucine are associated with alterations in enzymes involved in branched-chain amino acid metabolism. Am J Physiol Endocrinol Metab. 2007;293:E1552–63.PubMedCentralPubMed She P, Van Horn C, Reid T, Hutson SM, Cooney RN, Lynch CJ. Obesity-related elevations in plasma leucine are associated with alterations in enzymes involved in branched-chain amino acid metabolism. Am J Physiol Endocrinol Metab. 2007;293:E1552–63.PubMedCentralPubMed
67.
Zurück zum Zitat Newgard CB, An J, Bain J, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–26.PubMedCentralPubMed Newgard CB, An J, Bain J, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 2009;9:311–26.PubMedCentralPubMed
68.
Zurück zum Zitat Morris C, O’Grada C, Ryan M, Roche HM, Gibney MJ, Gibney ER, et al. The relationship between BMI and metabolomic profiles: a focus on amino acids. Proc Nutr Soc. 2012;71:634–8.PubMed Morris C, O’Grada C, Ryan M, Roche HM, Gibney MJ, Gibney ER, et al. The relationship between BMI and metabolomic profiles: a focus on amino acids. Proc Nutr Soc. 2012;71:634–8.PubMed
69.
Zurück zum Zitat McCormack SE, Shaham O, McCarthy MA, Deik AA, Wang TJ, Gerszten RE, et al. Circulating branched-chain amino acid concentrations are associated with obesity and future insulin resistance in children and adolescents. Pediatr Obes. 2013;8:52–61.PubMedCentralPubMed McCormack SE, Shaham O, McCarthy MA, Deik AA, Wang TJ, Gerszten RE, et al. Circulating branched-chain amino acid concentrations are associated with obesity and future insulin resistance in children and adolescents. Pediatr Obes. 2013;8:52–61.PubMedCentralPubMed
70.
Zurück zum Zitat Loridan L, Sadeghi-Nejad A, Senior B. Hypersecretion of insulin after the administration of L-leucine to obese children. J Pediatr. 1971;78:53–8.PubMed Loridan L, Sadeghi-Nejad A, Senior B. Hypersecretion of insulin after the administration of L-leucine to obese children. J Pediatr. 1971;78:53–8.PubMed
71.
Zurück zum Zitat Zhou YP, Grill VE. Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest. 1994;93:870–6.PubMedCentralPubMed Zhou YP, Grill VE. Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest. 1994;93:870–6.PubMedCentralPubMed
72.
Zurück zum Zitat Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, et al. Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest. 1999;103:253–9.PubMedCentralPubMed Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, et al. Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest. 1999;103:253–9.PubMedCentralPubMed
73.
Zurück zum Zitat Unger RH, Clark GO, Scherer PE, Orci L. Lipid homeostasis, lipotoxicity and the metabolic syndrome. Biochim Biophys Acta. 2010;1801:209–14.PubMed Unger RH, Clark GO, Scherer PE, Orci L. Lipid homeostasis, lipotoxicity and the metabolic syndrome. Biochim Biophys Acta. 2010;1801:209–14.PubMed
74.
Zurück zum Zitat Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, et al. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature. 2004;431:200–5.PubMed Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, et al. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature. 2004;431:200–5.PubMed
75.
Zurück zum Zitat Um SH, D’Alessio D, Thomas G. Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1. Cell Metab. 2006;3:393–402.PubMed Um SH, D’Alessio D, Thomas G. Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1. Cell Metab. 2006;3:393–402.PubMed
76.
Zurück zum Zitat Hoppe C, Mølgaard C, Michaelsen KF. Cow’s milk and linear growth in industrialized and developing countries. Ann Rev Nutr. 2006;26:131–73. Hoppe C, Mølgaard C, Michaelsen KF. Cow’s milk and linear growth in industrialized and developing countries. Ann Rev Nutr. 2006;26:131–73.
77.
Zurück zum Zitat Wiley AS. Dairy and milk consumption and child growth: Is BMI involved? An analysis of NHANES 1999–2004. Am J Hum Biol. 2010;22:517–25.PubMed Wiley AS. Dairy and milk consumption and child growth: Is BMI involved? An analysis of NHANES 1999–2004. Am J Hum Biol. 2010;22:517–25.PubMed
78.
Zurück zum Zitat Berkey CS, Rocket HR, Willet WC, Colditz GA. Milk, dairy fat, dietary calcium, and weight gain. Arch Pediatr Adolesc Med. 2005;159:543–50.PubMed Berkey CS, Rocket HR, Willet WC, Colditz GA. Milk, dairy fat, dietary calcium, and weight gain. Arch Pediatr Adolesc Med. 2005;159:543–50.PubMed
79.
Zurück zum Zitat Matthews VL, Wien M, Sabaté J. The risk of child and adolescent overweight is related to types of food consumed. Nutr J. 2011;10:71.PubMedCentralPubMed Matthews VL, Wien M, Sabaté J. The risk of child and adolescent overweight is related to types of food consumed. Nutr J. 2011;10:71.PubMedCentralPubMed
80.
Zurück zum Zitat Arnberg K, Mølgaard C, Michaelsen KF, Jensen SM, Trolle E, Larnkjær A. Skim milk, whey, and casein increase body weight and whey and casein increase plasma C-peptide concentration in overweight adolescents. J Nutr. 2012;142:2083–90.PubMed Arnberg K, Mølgaard C, Michaelsen KF, Jensen SM, Trolle E, Larnkjær A. Skim milk, whey, and casein increase body weight and whey and casein increase plasma C-peptide concentration in overweight adolescents. J Nutr. 2012;142:2083–90.PubMed
81.
Zurück zum Zitat Barr SI, McCarron DA, Heaney RP, Dawson-Hughes B, Berga SL, Stern JS, et al. Effects of increased consumption of fluid milk on energy and nutrient intake, body weight, and cardiovascular risk factors in healthy older adults. Am J Diet Assoc. 2000;100:810–7. Barr SI, McCarron DA, Heaney RP, Dawson-Hughes B, Berga SL, Stern JS, et al. Effects of increased consumption of fluid milk on energy and nutrient intake, body weight, and cardiovascular risk factors in healthy older adults. Am J Diet Assoc. 2000;100:810–7.
82.
Zurück zum Zitat Chen M, Pan A, Malik VS, Hu FB. Effects of dairy intake on body weight and fat: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2012;96:735–47.PubMedCentralPubMed Chen M, Pan A, Malik VS, Hu FB. Effects of dairy intake on body weight and fat: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2012;96:735–47.PubMedCentralPubMed
83.
Zurück zum Zitat Abreu S, Santos R, Moreira C, Vale S, Santos PC, Soares-Miranda L, et al. Association between dairy product intake and abdominal obesity in Azorean adolescents. Eur J Clin Nutr. 2012;66:830–5.PubMed Abreu S, Santos R, Moreira C, Vale S, Santos PC, Soares-Miranda L, et al. Association between dairy product intake and abdominal obesity in Azorean adolescents. Eur J Clin Nutr. 2012;66:830–5.PubMed
84.
Zurück zum Zitat Abreu S, Santos R, Moreira C, Santos PC, Vale S, Soares-Miranda L, et al. Milk intake is inversely related to body mass index and body fat in girls. Eur J Pediatr. 2012;171:1467–74.PubMed Abreu S, Santos R, Moreira C, Santos PC, Vale S, Soares-Miranda L, et al. Milk intake is inversely related to body mass index and body fat in girls. Eur J Pediatr. 2012;171:1467–74.PubMed
85.
Zurück zum Zitat Männik J, Vaas P, Rull K, Teesalu P, Laan M. Differential placental expression profile of human growth hormone/chorionic somatomammotropin genes in pregnancies with pre-eclampsia and gestational diabetes mellitus. Mol Cell Endocrinol. 2012;355:180–7.PubMedCentralPubMed Männik J, Vaas P, Rull K, Teesalu P, Laan M. Differential placental expression profile of human growth hormone/chorionic somatomammotropin genes in pregnancies with pre-eclampsia and gestational diabetes mellitus. Mol Cell Endocrinol. 2012;355:180–7.PubMedCentralPubMed
86.
Zurück zum Zitat Olafsdottir AS, Skuladottir GV, Thorsdottir I, Hauksson A, Steingrimsdottir L. Maternal diet in early and late pregnancy in relation to weight gain. Int J Obes (Lond). 2006;30:492–9. Olafsdottir AS, Skuladottir GV, Thorsdottir I, Hauksson A, Steingrimsdottir L. Maternal diet in early and late pregnancy in relation to weight gain. Int J Obes (Lond). 2006;30:492–9.
87.
Zurück zum Zitat Olsen SF, Halldorsson TI, Willett WC, Knudsen VK, Gillman MW, Mikkelsen TB, et al. Milk consumption during pregnancy is associated with increased infant size at birth: prospective cohort study. Am J Clin Nutr. 2007;86:1104–10.PubMed Olsen SF, Halldorsson TI, Willett WC, Knudsen VK, Gillman MW, Mikkelsen TB, et al. Milk consumption during pregnancy is associated with increased infant size at birth: prospective cohort study. Am J Clin Nutr. 2007;86:1104–10.PubMed
88.
Zurück zum Zitat Rao S, Yajnik CS, Kanade A, Fall CH, Margetts BM, Jackson AA, et al. Intake of micronutrient-rich foods in rural Indian mothers is associated with the size of their babies at birth: Pune Maternal Nutrition Study. J Nutr. 2001;131:1217–24.PubMed Rao S, Yajnik CS, Kanade A, Fall CH, Margetts BM, Jackson AA, et al. Intake of micronutrient-rich foods in rural Indian mothers is associated with the size of their babies at birth: Pune Maternal Nutrition Study. J Nutr. 2001;131:1217–24.PubMed
89.
Zurück zum Zitat Cramer DW, Beck P, Makowski EL. Correlation of gestational age with maternal human chorionic somatomammotropin and maternal and fetal growth hormone plasma concentrations during labor. Am J Obstet Gynecol. 1971;109:649–55.PubMed Cramer DW, Beck P, Makowski EL. Correlation of gestational age with maternal human chorionic somatomammotropin and maternal and fetal growth hormone plasma concentrations during labor. Am J Obstet Gynecol. 1971;109:649–55.PubMed
90.
Zurück zum Zitat Lindberg BS, Nilsson BA. Human placental lactogen (HPL) levels in abnormal pregnancies. J Obstet Gynaecol Br Commonw. 1973;80:1046–53.PubMed Lindberg BS, Nilsson BA. Human placental lactogen (HPL) levels in abnormal pregnancies. J Obstet Gynaecol Br Commonw. 1973;80:1046–53.PubMed
91.
Zurück zum Zitat Sciarra JJ, Sherwood LM, Varma AA, Lundberg WB. Human placental lactogen (HPL) and placental weight. Am J Obstet Gynecol. 1968;101:413–6.PubMed Sciarra JJ, Sherwood LM, Varma AA, Lundberg WB. Human placental lactogen (HPL) and placental weight. Am J Obstet Gynecol. 1968;101:413–6.PubMed
92.
Zurück zum Zitat Boyce A, Schwartz D, Hubert C, Cedard L, Dreyfus J. Smoking, human placental lactogen and birth weight. Br J Obstet Gynaecol. 1975;82:964–7.PubMed Boyce A, Schwartz D, Hubert C, Cedard L, Dreyfus J. Smoking, human placental lactogen and birth weight. Br J Obstet Gynaecol. 1975;82:964–7.PubMed
93.
Zurück zum Zitat Letchworth AT, Boardman RJ, Bristow C, Landon J, Chard T. A rapid semi-automated method for the measurement of human chorionic sommatomammotrophin. The normal range in the third trimester and its relation to fetal weight. J Obstet Gynaecol Br Commonw. 1971;78:542–8.PubMed Letchworth AT, Boardman RJ, Bristow C, Landon J, Chard T. A rapid semi-automated method for the measurement of human chorionic sommatomammotrophin. The normal range in the third trimester and its relation to fetal weight. J Obstet Gynaecol Br Commonw. 1971;78:542–8.PubMed
94.
Zurück zum Zitat Lindberg BS, Nilsson BA. Variations in maternal plasma levels of human placental lactogen (HPL) in normal pregnancy and labour. J Obstet Gynaecol Br Commonw. 1973;80:619–26.PubMed Lindberg BS, Nilsson BA. Variations in maternal plasma levels of human placental lactogen (HPL) in normal pregnancy and labour. J Obstet Gynaecol Br Commonw. 1973;80:619–26.PubMed
95.
Zurück zum Zitat Freemark M. Placental hormones and the control of fetal growth. J Clin Endocrinol Metab. 2010;95:2054–7.PubMed Freemark M. Placental hormones and the control of fetal growth. J Clin Endocrinol Metab. 2010;95:2054–7.PubMed
96.
Zurück zum Zitat Henleigh PA, Cheatum SG, Spellacy WN. Oxytocinase and human placental lactogen for the prediction of intrauterine growth retardation. Am J Obstet Gynaecol. 1977;129:675–8. Henleigh PA, Cheatum SG, Spellacy WN. Oxytocinase and human placental lactogen for the prediction of intrauterine growth retardation. Am J Obstet Gynaecol. 1977;129:675–8.
97.
98.
Zurück zum Zitat Larqué E, Riuz-Palacios M, Koletzko B. Placental regulation of fetal nutrient supply. Curr Opin Clin Nutr Metab Care. 2013;16:292–7.PubMed Larqué E, Riuz-Palacios M, Koletzko B. Placental regulation of fetal nutrient supply. Curr Opin Clin Nutr Metab Care. 2013;16:292–7.PubMed
99.
Zurück zum Zitat Roos S, Jansson N, Palmberg I, Säljö K, Powell TL, Jansson T. Mammalian target of rapamycin in the human placenta regulates leucine transport and is down-regulated in restricted growth. J Physiol. 2007;582:449–59.PubMedCentralPubMed Roos S, Jansson N, Palmberg I, Säljö K, Powell TL, Jansson T. Mammalian target of rapamycin in the human placenta regulates leucine transport and is down-regulated in restricted growth. J Physiol. 2007;582:449–59.PubMedCentralPubMed
100.
Zurück zum Zitat Jansson T, Aye IL, Goberdhan DC. The emerging role of mTORC1 signaling in placental nutrient-sensing. Placenta. 2012;33 Suppl 2:e23–9.PubMedCentralPubMed Jansson T, Aye IL, Goberdhan DC. The emerging role of mTORC1 signaling in placental nutrient-sensing. Placenta. 2012;33 Suppl 2:e23–9.PubMedCentralPubMed
101.
Zurück zum Zitat Roos S, Lagerlöf O, Wennergren M, Powell TL, Jansson T. Regulation of amino acid transporters by glucose and growth factors in cultured primary human trophoblast cells is mediated by mTOR signaling. Am J Physiol Cell Physiol. 2009;297:C723–31.PubMed Roos S, Lagerlöf O, Wennergren M, Powell TL, Jansson T. Regulation of amino acid transporters by glucose and growth factors in cultured primary human trophoblast cells is mediated by mTOR signaling. Am J Physiol Cell Physiol. 2009;297:C723–31.PubMed
102.
Zurück zum Zitat Jansson N, Rosario FJ, Gaccioli F, Lager S, Jones HN, Roos S, et al. Activation of placental mTOR signaling and amino acid transporters in obese women giving birth to large babies. J Clin Endocrinol Metab. 2013;98:105–13.PubMedCentralPubMed Jansson N, Rosario FJ, Gaccioli F, Lager S, Jones HN, Roos S, et al. Activation of placental mTOR signaling and amino acid transporters in obese women giving birth to large babies. J Clin Endocrinol Metab. 2013;98:105–13.PubMedCentralPubMed
103.
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.PubMed 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.PubMed
104.
Zurück zum Zitat Lu Y, Qian L, Zhang Q, Chen B, Gui L, Huang D, et al. Palmitate induces apoptosis in mouse aortic endothelial cells and endothelial dysfunction in mice fed high-calorie and high-cholesterol diets. Life Sci. 2013;92:1165–73.PubMed Lu Y, Qian L, Zhang Q, Chen B, Gui L, Huang D, et al. Palmitate induces apoptosis in mouse aortic endothelial cells and endothelial dysfunction in mice fed high-calorie and high-cholesterol diets. Life Sci. 2013;92:1165–73.PubMed
105.
Zurück zum Zitat Kavitha JV, Rosario FJ, Nijland MJ, McDonald TJ, Wu G, Kanai Y, et al. Down-regulation of placental mTOR, insulin/IGF-I signaling, and nutrient transporters in response to maternal nutrient restriction in the baboon. FASEB J. 2014;28:1294–2305.PubMed Kavitha JV, Rosario FJ, Nijland MJ, McDonald TJ, Wu G, Kanai Y, et al. Down-regulation of placental mTOR, insulin/IGF-I signaling, and nutrient transporters in response to maternal nutrient restriction in the baboon. FASEB J. 2014;28:1294–2305.PubMed
106.
Zurück zum Zitat Newbern D, Freemark M. Placental hormones and the control of maternal metabolism and fetal growth. Curr Opin Endocrinol Diabetes Obes. 2011;18:409–16.PubMed Newbern D, Freemark M. Placental hormones and the control of maternal metabolism and fetal growth. Curr Opin Endocrinol Diabetes Obes. 2011;18:409–16.PubMed
107.
Zurück zum Zitat Hennighausen L, Robibson GW. Interpretation of cytokine signalling through the transcription factors STAT5A and STAT5B. Genes Dev. 2008;22:711–21.PubMedCentralPubMed Hennighausen L, Robibson GW. Interpretation of cytokine signalling through the transcription factors STAT5A and STAT5B. Genes Dev. 2008;22:711–21.PubMedCentralPubMed
108.
Zurück zum Zitat Cao J, Gowri PM, Ganguly TC, Wood M, Hyde JF, Talamantes F, et al. PRL, placental lactogen, and GH induce NA(+)/taurocholate-cotransporting polypeptide gene expression by activating signal transducer and activator of transcription-5 in liver cells. Endocrinology. 2001;142:4212–22.PubMed Cao J, Gowri PM, Ganguly TC, Wood M, Hyde JF, Talamantes F, et al. PRL, placental lactogen, and GH induce NA(+)/taurocholate-cotransporting polypeptide gene expression by activating signal transducer and activator of transcription-5 in liver cells. Endocrinology. 2001;142:4212–22.PubMed
109.
Zurück zum Zitat Kondegowda NG, Mozar A, Chin C, Otero A, Garcia-Ocana A, Vasavada RC. Lactogens protect rodent and human beta cells agianst glucolipotoxicity-induced cell death through Janus kinase-2 (JAK2)/signal transducer and activator of transcription-5 (STAT5) signalling. Diabetologica. 2012;55:1721–32. Kondegowda NG, Mozar A, Chin C, Otero A, Garcia-Ocana A, Vasavada RC. Lactogens protect rodent and human beta cells agianst glucolipotoxicity-induced cell death through Janus kinase-2 (JAK2)/signal transducer and activator of transcription-5 (STAT5) signalling. Diabetologica. 2012;55:1721–32.
110.
Zurück zum Zitat Fujinaka Y, Takane K, Yamashita H, Vasavada RC. Lactogens promote beta cell survival through JAK2/STAT5 activation and Bcl-XL upregulation. J Biol Chem. 2007;282:30707–17.PubMed Fujinaka Y, Takane K, Yamashita H, Vasavada RC. Lactogens promote beta cell survival through JAK2/STAT5 activation and Bcl-XL upregulation. J Biol Chem. 2007;282:30707–17.PubMed
111.
Zurück zum Zitat Pedersen NG, Juul A, Chrisitansen M, Wojdemann KR, Tabor A. Maternal serum placental growth hormone, but not human placental lactogen or insulin growth factor-1, is positively associated with fetal growth in the first half of pregnancy. Ultrasound Obstet Gynecol. 2010;36:534–41.PubMed Pedersen NG, Juul A, Chrisitansen M, Wojdemann KR, Tabor A. Maternal serum placental growth hormone, but not human placental lactogen or insulin growth factor-1, is positively associated with fetal growth in the first half of pregnancy. Ultrasound Obstet Gynecol. 2010;36:534–41.PubMed
112.
Zurück zum Zitat Howard JK, Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins. Trends Endocrinol Metab. 2006;17:365–71.PubMed Howard JK, Flier JS. Attenuation of leptin and insulin signaling by SOCS proteins. Trends Endocrinol Metab. 2006;17:365–71.PubMed
113.
Zurück zum Zitat Lebrun P, Van Obberghen E. SOCS proteins causing trouble in insulin action. Acta Physiol (Oxf). 2008;192:29–36. Lebrun P, Van Obberghen E. SOCS proteins causing trouble in insulin action. Acta Physiol (Oxf). 2008;192:29–36.
114.
Zurück zum Zitat Lebrun P, Cognard E, Gontard P, Bellon-Paul R, Filloux C, Berthault MF, et al. The suppressor of cytokine signalling 2 (SOCS2) is a key repressor of insulin secretion. Diabetologia. 2010;53:1935–46.PubMed Lebrun P, Cognard E, Gontard P, Bellon-Paul R, Filloux C, Berthault MF, et al. The suppressor of cytokine signalling 2 (SOCS2) is a key repressor of insulin secretion. Diabetologia. 2010;53:1935–46.PubMed
115.
Zurück zum Zitat Yang Z, Hulver M, McMillan RP, Cai L, Kershaw EE, Yu L, et al. Regulation of insulin and leptin signaling by muscle suppressor of cytokine signaling 3 (SOCS3). PLoS One. 2012;7:e47493.PubMedCentralPubMed Yang Z, Hulver M, McMillan RP, Cai L, Kershaw EE, Yu L, et al. Regulation of insulin and leptin signaling by muscle suppressor of cytokine signaling 3 (SOCS3). PLoS One. 2012;7:e47493.PubMedCentralPubMed
116.
Zurück zum Zitat Dann SG, Selvaraj A, Thomas G. mTOR Complex1-S6K1 signaling: at the crossroads of obesity, diabetes and cancer. Trends Mol Med. 2007;13:252–9.PubMed Dann SG, Selvaraj A, Thomas G. mTOR Complex1-S6K1 signaling: at the crossroads of obesity, diabetes and cancer. Trends Mol Med. 2007;13:252–9.PubMed
117.
Zurück zum Zitat Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012;441:1–21.PubMed Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012;441:1–21.PubMed
118.
Zurück zum Zitat Patti ME, Brambilla E, Luzi L, Landaker EJ, Kahn CR. Bidirectional modulation of insulin action by amino acids. J Clin Invest. 1998;101:1519–29.PubMedCentralPubMed Patti ME, Brambilla E, Luzi L, Landaker EJ, Kahn CR. Bidirectional modulation of insulin action by amino acids. J Clin Invest. 1998;101:1519–29.PubMedCentralPubMed
119.
Zurück zum Zitat Krebs M, Krssak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, et al. Mechanism of amino acid-induced skeletal muscle insulin resistance in humans. Diabetes. 2002;51:599–605.PubMed Krebs M, Krssak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, et al. Mechanism of amino acid-induced skeletal muscle insulin resistance in humans. Diabetes. 2002;51:599–605.PubMed
120.
Zurück zum Zitat Tremblay F, Krebs M, Dombrowski L, Brehm A, Bernroider E, Roth E, et al. Overactivation of S6 kinase 1 as a cause of human insulin resistance during increased amino acid availability. Diabetes. 2005;54:2674–84.PubMed Tremblay F, Krebs M, Dombrowski L, Brehm A, Bernroider E, Roth E, et al. Overactivation of S6 kinase 1 as a cause of human insulin resistance during increased amino acid availability. Diabetes. 2005;54:2674–84.PubMed
121.
Zurück zum Zitat Tremblay F, Brulé S, Hee Um S, Masuda K, Roden M, Sun XJ, et al. Identification of Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance. Proc Natl Acad Sci U S A. 2007;104:14056–61.PubMedCentralPubMed Tremblay F, Brulé S, Hee Um S, Masuda K, Roden M, Sun XJ, et al. Identification of Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance. Proc Natl Acad Sci U S A. 2007;104:14056–61.PubMedCentralPubMed
122.
Zurück zum Zitat Shah OJ, Wang Z, Hunter T. Inappropriate activation of the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol. 2004;14:1650–6.PubMed Shah OJ, Wang Z, Hunter T. Inappropriate activation of the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol. 2004;14:1650–6.PubMed
123.
Zurück zum Zitat Lu J, Xie G, Jia W, Jia W. Insulin resistance and the metabolism of branched-chain amino acids. Front Med. 2013;7:53–9.PubMed Lu J, Xie G, Jia W, Jia W. Insulin resistance and the metabolism of branched-chain amino acids. Front Med. 2013;7:53–9.PubMed
124.
Zurück zum Zitat Zeng M, Che Z, Liang Y, Wang B, Chen X, Li H, et al. GC-MS based plasma metabolic profiling of type 2 diabetes mellitus. Chromatographia. 2009;69:941–8. Zeng M, Che Z, Liang Y, Wang B, Chen X, Li H, et al. GC-MS based plasma metabolic profiling of type 2 diabetes mellitus. Chromatographia. 2009;69:941–8.
125.
Zurück zum Zitat Iglesias P, Selgas R, Romero S, Díez JJ. Biological role, clinical significance, and therapeutic possibilities of the recently discovered metabolic hormone fibroblastic growth factor 21. Eur J Endocrinol. 2012;167:301–9.PubMed Iglesias P, Selgas R, Romero S, Díez JJ. Biological role, clinical significance, and therapeutic possibilities of the recently discovered metabolic hormone fibroblastic growth factor 21. Eur J Endocrinol. 2012;167:301–9.PubMed
126.
Zurück zum Zitat Zhang X, Yeung DC, Karpisek M, Stejskal D, Zhou ZG, Liu F, et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes. 2008;57:1246–53.PubMed Zhang X, Yeung DC, Karpisek M, Stejskal D, Zhou ZG, Liu F, et al. Serum FGF21 levels are increased in obesity and are independently associated with the metabolic syndrome in humans. Diabetes. 2008;57:1246–53.PubMed
127.
Zurück zum Zitat Tan BK, Sivakumar K, Bari MF, Vatish M, Randeva HS. Lower cerebrospinal fluid/plasma fibroblast growth factor 21 (FGF21) ratios and placental FGF21 production in gestational diabetes. PLoS One. 2013;8:e65254.PubMedCentralPubMed Tan BK, Sivakumar K, Bari MF, Vatish M, Randeva HS. Lower cerebrospinal fluid/plasma fibroblast growth factor 21 (FGF21) ratios and placental FGF21 production in gestational diabetes. PLoS One. 2013;8:e65254.PubMedCentralPubMed
128.
Zurück zum Zitat Dekker Nitert M, Barrett HL, Kubala MH, Scholz Romero K, Denny KJ, Woodruff TM, et al. Increased placental expression of fibroblast growth factor 21 in gestational diabetes mellitus. J Clin Endocrinol Metab. 2014;99:E591–8.PubMed Dekker Nitert M, Barrett HL, Kubala MH, Scholz Romero K, Denny KJ, Woodruff TM, et al. Increased placental expression of fibroblast growth factor 21 in gestational diabetes mellitus. J Clin Endocrinol Metab. 2014;99:E591–8.PubMed
129.
Zurück zum Zitat Yu J, Zhao L, Wang A, Eleswarapu S, Ge X, Chen D, et al. Growth hormone stimulates transcription of the fibroblast growth factor 21 gene in the liver through the signal transducer and activator of transcription 5. Endocrinology. 2012;153:750–8.PubMed Yu J, Zhao L, Wang A, Eleswarapu S, Ge X, Chen D, et al. Growth hormone stimulates transcription of the fibroblast growth factor 21 gene in the liver through the signal transducer and activator of transcription 5. Endocrinology. 2012;153:750–8.PubMed
130.
Zurück zum Zitat Cui Y, Giesy SL, Hassan M, Davis K, Zhao S, Boisclair YR. Hepatic FGF21 production is increased in late pregnancy in the mouse. Am J Physiol Regul Integr Comp Physiol. 2014;307:R290–8.PubMed Cui Y, Giesy SL, Hassan M, Davis K, Zhao S, Boisclair YR. Hepatic FGF21 production is increased in late pregnancy in the mouse. Am J Physiol Regul Integr Comp Physiol. 2014;307:R290–8.PubMed
131.
Zurück zum Zitat Cornu M, Oppliger W, Albert V, Robitaille AM, Trapani F, Quagliata L, et al. Hepatic mTORC1 controls locomotor activity, body temperature and lipid metabolism through FGF21. Proc Natl Acad Sci U S A. 2014;111:11592–9.PubMed Cornu M, Oppliger W, Albert V, Robitaille AM, Trapani F, Quagliata L, et al. Hepatic mTORC1 controls locomotor activity, body temperature and lipid metabolism through FGF21. Proc Natl Acad Sci U S A. 2014;111:11592–9.PubMed
132.
Zurück zum Zitat Li K, Li L, Yang M, Liu H, Boden G, Yang G. The effects of fibroblast growth factor-21 knockdown and over-expression on its signaling pathway and glucose-lipid metabolism in vitro. Mol Cell Endocrinol. 2012;348:21–6.PubMed Li K, Li L, Yang M, Liu H, Boden G, Yang G. The effects of fibroblast growth factor-21 knockdown and over-expression on its signaling pathway and glucose-lipid metabolism in vitro. Mol Cell Endocrinol. 2012;348:21–6.PubMed
133.
Zurück zum Zitat Ericsson A, Hamark B, Powell TL, Jansson T. Glucose transporter isoform 4 is expressed in the syncytiotrophoblast of first trimester human placenta. Hum Reprod. 2005;20:521–30.PubMed Ericsson A, Hamark B, Powell TL, Jansson T. Glucose transporter isoform 4 is expressed in the syncytiotrophoblast of first trimester human placenta. Hum Reprod. 2005;20:521–30.PubMed
134.
Zurück zum Zitat Jansson T, Wennergren M, Illsley NP. Glucose transporter protein expression in human placenta throughout gestation and in intrauterine growth retardation. J Clin Endocrinol Metab. 1993;77:1554–62.PubMed Jansson T, Wennergren M, Illsley NP. Glucose transporter protein expression in human placenta throughout gestation and in intrauterine growth retardation. J Clin Endocrinol Metab. 1993;77:1554–62.PubMed
135.
Zurück zum Zitat Jansson T, Wennergren M, Powell TL. Placental glucose transport and GLUT1 expression in insulin-dependent diabetes. Am J Obstet Gynecol. 1999;180:163–8.PubMed Jansson T, Wennergren M, Powell TL. Placental glucose transport and GLUT1 expression in insulin-dependent diabetes. Am J Obstet Gynecol. 1999;180:163–8.PubMed
136.
Zurück zum Zitat Gaither K, Quraishi AN, Illsley NP. Diabetes alters the expression and activity of the human placental GLUT1 glucose transporter. J Clin Endocrinol Metab. 1999;84:695–701.PubMed Gaither K, Quraishi AN, Illsley NP. Diabetes alters the expression and activity of the human placental GLUT1 glucose transporter. J Clin Endocrinol Metab. 1999;84:695–701.PubMed
137.
Zurück zum Zitat Acosta O, Ramirez VI, Lager S, Gaccioli F, Dudley DJ, Powell TL, et al. Increased glucose and placental GLUT-1 in large babies of obese non-diabetic mothers. Am J Obstet Gynecol. 2014; Aug 14 [Epub ahead of print]. Acosta O, Ramirez VI, Lager S, Gaccioli F, Dudley DJ, Powell TL, et al. Increased glucose and placental GLUT-1 in large babies of obese non-diabetic mothers. Am J Obstet Gynecol. 2014; Aug 14 [Epub ahead of print].
138.
Zurück zum Zitat Jiang H, Wu W, Zhang M, Li J, Peng Y, Miao TT, et al. Aberrant upregulation of miR-21 in placental tissues of macrosomia. J Perinatol. 2014;34:658–63.PubMed Jiang H, Wu W, Zhang M, Li J, Peng Y, Miao TT, et al. Aberrant upregulation of miR-21 in placental tissues of macrosomia. J Perinatol. 2014;34:658–63.PubMed
139.
Zurück zum Zitat Chen X, Gao C, Li H, Huang L, Sun Q, Dong Y, et al. Identification and characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products. Cell Res. 2010;20:1128–37.PubMed Chen X, Gao C, Li H, Huang L, Sun Q, Dong Y, et al. Identification and characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products. Cell Res. 2010;20:1128–37.PubMed
140.
Zurück zum Zitat Baier SR, Nguyen C, Xie F, Wood JR, Zempleni J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr. 2014;144:1495–500.PubMed Baier SR, Nguyen C, Xie F, Wood JR, Zempleni J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr. 2014;144:1495–500.PubMed
141.
Zurück zum Zitat Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133:647–58.PubMedCentralPubMed Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133:647–58.PubMedCentralPubMed
142.
Zurück zum Zitat Han M, Liu M, Wang Y, Chen X, Xu J, Sun Y, et al. Antagonism of miR-21 reverses epithelial-mesenchymal transition and cancer stem cell phenotype through AKT/ERK1/2 inactivation by targeting PTEN. PLoS One. 2012;7:e39520.PubMedCentralPubMed Han M, Liu M, Wang Y, Chen X, Xu J, Sun Y, et al. Antagonism of miR-21 reverses epithelial-mesenchymal transition and cancer stem cell phenotype through AKT/ERK1/2 inactivation by targeting PTEN. PLoS One. 2012;7:e39520.PubMedCentralPubMed
143.
Zurück zum Zitat Dey N, Das F, Mariappan MM, Mandal CC, Ghosh-Choudhury N, Kasinath BS, et al. MicroRNA-21 orchestrates high glucose-induced signals to TOR complex 1, resulting in renal cell pathology in diabetes. J Biol Chem. 2011;286:25586–603.PubMedCentralPubMed Dey N, Das F, Mariappan MM, Mandal CC, Ghosh-Choudhury N, Kasinath BS, et al. MicroRNA-21 orchestrates high glucose-induced signals to TOR complex 1, resulting in renal cell pathology in diabetes. J Biol Chem. 2011;286:25586–603.PubMedCentralPubMed
144.
Zurück zum Zitat Dey N, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. TGFβ-stimulated microRNA-21 utilizes PTEN to orchestrate AKT/mTORC1 signaling for mesangial cell hypertrophy and matrix expansion. PLoS One. 2012;7:e42316.PubMedCentralPubMed Dey N, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. TGFβ-stimulated microRNA-21 utilizes PTEN to orchestrate AKT/mTORC1 signaling for mesangial cell hypertrophy and matrix expansion. PLoS One. 2012;7:e42316.PubMedCentralPubMed
145.
Zurück zum Zitat Sayed D, Rane S, Lypowy J, He M, Chen IY, Vashistha H, et al. MicroRNA-21 targets Sprouty2 and promotes cellular outgrowths. Mol Biol Cell. 2008;19:3272–82.PubMedCentralPubMed Sayed D, Rane S, Lypowy J, He M, Chen IY, Vashistha H, et al. MicroRNA-21 targets Sprouty2 and promotes cellular outgrowths. Mol Biol Cell. 2008;19:3272–82.PubMedCentralPubMed
146.
Zurück zum Zitat Dariminpourain M, Wang S, Ittmann M, Kwabi-Addo B. Transcriptional and post-transcriptional regulation of Sprouty1, a receptor tyrosine kinase inhibitor in prostate cancer. Prostate Cancer Prostatic Dis. 2011;14:279–85. Dariminpourain M, Wang S, Ittmann M, Kwabi-Addo B. Transcriptional and post-transcriptional regulation of Sprouty1, a receptor tyrosine kinase inhibitor in prostate cancer. Prostate Cancer Prostatic Dis. 2011;14:279–85.
147.
Zurück zum Zitat Frey MR, Carraro G, Batra RK, Polk DB, Warburton D. Sprouty keeps bowel kinases regular in colon cancer, while miR-21 targets Sprouty. Cancer Biol Ther. 2011;11:122–4.PubMed Frey MR, Carraro G, Batra RK, Polk DB, Warburton D. Sprouty keeps bowel kinases regular in colon cancer, while miR-21 targets Sprouty. Cancer Biol Ther. 2011;11:122–4.PubMed
148.
Zurück zum Zitat Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27:2128–36.PubMed Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27:2128–36.PubMed
149.
Zurück zum Zitat Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH, et al. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 2008;27:4373–9.PubMed Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH, et al. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 2008;27:4373–9.PubMed
150.
Zurück zum Zitat Carayol N, Katsoulidis E, Sassano A, Altman JK, Druker BJ, Platanias LC. Suppression of programmed cell death 4 (PDCD4) protein expression by BCR-ABL-regulated engagement of the mTOR/p70 S6 kinase pathway. J Biol Chem. 2008;28:8601–10. Carayol N, Katsoulidis E, Sassano A, Altman JK, Druker BJ, Platanias LC. Suppression of programmed cell death 4 (PDCD4) protein expression by BCR-ABL-regulated engagement of the mTOR/p70 S6 kinase pathway. J Biol Chem. 2008;28:8601–10.
151.
Zurück zum Zitat Ng R, Song G, Roll GR, Frandsen NM, Willenbring H. A microRNA-21 surge facilitates rapid cyclin D1 translation and cell cycle progression in mouse liver regeneration. J Clin Invest. 2012;122:1097–108.PubMedCentralPubMed Ng R, Song G, Roll GR, Frandsen NM, Willenbring H. A microRNA-21 surge facilitates rapid cyclin D1 translation and cell cycle progression in mouse liver regeneration. J Clin Invest. 2012;122:1097–108.PubMedCentralPubMed
152.
Zurück zum Zitat Dennis MD, Jefferson LS, Kimball SR. Role of p70S6K1-mediated phosphorylation of eIF4B and PDCD4 proteins in the regulation of protein synthesis. J Biol Chem. 2012;287:42890–9.PubMedCentralPubMed Dennis MD, Jefferson LS, Kimball SR. Role of p70S6K1-mediated phosphorylation of eIF4B and PDCD4 proteins in the regulation of protein synthesis. J Biol Chem. 2012;287:42890–9.PubMedCentralPubMed
153.
Zurück zum Zitat Kim YJ, Hwang SJ, Bae YC, Jung JS. MiR-21 regulates adipogenic differentiation through the modulation of TGF-beta signaling in mesenchymal stem cells derived from human adipose tissue. Stem Cells. 2009;27:3093–102.PubMed Kim YJ, Hwang SJ, Bae YC, Jung JS. MiR-21 regulates adipogenic differentiation through the modulation of TGF-beta signaling in mesenchymal stem cells derived from human adipose tissue. Stem Cells. 2009;27:3093–102.PubMed
154.
Zurück zum Zitat Kim YJ, Hwang SH, Cho HH, Shin KK, Bae YC, Jung JS. MicroRNA 21 regulates the proliferation of human adipose tissue-derived mesenchymal stem cells and high-fat diet-induced obesity alters microRNA 21 expression in white adipose tissues. J Cell Physiol. 2012;227:183–93.PubMed Kim YJ, Hwang SH, Cho HH, Shin KK, Bae YC, Jung JS. MicroRNA 21 regulates the proliferation of human adipose tissue-derived mesenchymal stem cells and high-fat diet-induced obesity alters microRNA 21 expression in white adipose tissues. J Cell Physiol. 2012;227:183–93.PubMed
155.
Zurück zum Zitat Seeger T, Fischer A, Muhly-Reinholz M, Zeiher AM, Dimmeler S. Long-term inhibition of miR-21 leads to reduction of obesity in db/db mice. Obesity (Silver Spring). 2014;22:2352–60. Seeger T, Fischer A, Muhly-Reinholz M, Zeiher AM, Dimmeler S. Long-term inhibition of miR-21 leads to reduction of obesity in db/db mice. Obesity (Silver Spring). 2014;22:2352–60.
156.
Zurück zum Zitat Heppe DH, van Dam RM, Willemsen SP, den Breeijen H, Raat H, Hofman A, et al. Maternal milk consumption, fetal growth, and the risks of neonatal complications: the Generation R Study. Am J Clin Nutr. 2011;94:501–9.PubMed Heppe DH, van Dam RM, Willemsen SP, den Breeijen H, Raat H, Hofman A, et al. Maternal milk consumption, fetal growth, and the risks of neonatal complications: the Generation R Study. Am J Clin Nutr. 2011;94:501–9.PubMed
157.
Zurück zum Zitat Hadlock FP, Harrist RB, Sharman RS, Deter RL, Park SK. Estimation of fetal weight with the use of head, body, and femur measurements - a prospective study. Am J Obstet Gynecol. 1985;151:333–7.PubMed Hadlock FP, Harrist RB, Sharman RS, Deter RL, Park SK. Estimation of fetal weight with the use of head, body, and femur measurements - a prospective study. Am J Obstet Gynecol. 1985;151:333–7.PubMed
158.
Zurück zum Zitat Ludvigsson JF, Ludvigsson J. Milk consumption during pregnancy and infant birthweight. Acta Paediatr. 2004;93:1474–8.PubMed Ludvigsson JF, Ludvigsson J. Milk consumption during pregnancy and infant birthweight. Acta Paediatr. 2004;93:1474–8.PubMed
159.
Zurück zum Zitat Mannion CA, Gray-Donald K, Koski KG. Association of low intake of milk and vitamin D during pregnancy with decreased birth weight. CMAJ. 2006;174:1273–7.PubMedCentralPubMed Mannion CA, Gray-Donald K, Koski KG. Association of low intake of milk and vitamin D during pregnancy with decreased birth weight. CMAJ. 2006;174:1273–7.PubMedCentralPubMed
160.
Zurück zum Zitat Moore VM, Davies MJ, Willson KJ, Worsley A, Robinson JS. Dietary composition of pregnant women is related to size of the baby at birth. J Nutr. 2004;134:1820–6.PubMed Moore VM, Davies MJ, Willson KJ, Worsley A, Robinson JS. Dietary composition of pregnant women is related to size of the baby at birth. J Nutr. 2004;134:1820–6.PubMed
161.
Zurück zum Zitat Chan GM, McElligott K, McNaught T, Gill G. Effects of dietary calcium intervention on adolescent mothers and newborns: A randomized controlled trial. Obstet Gynecol. 2006;108:565–71.PubMed Chan GM, McElligott K, McNaught T, Gill G. Effects of dietary calcium intervention on adolescent mothers and newborns: A randomized controlled trial. Obstet Gynecol. 2006;108:565–71.PubMed
162.
Zurück zum Zitat Brantsæter AL, Olafsdottir AS, Forsum E, Olsen SF, Thorsdottir I. Does milk and dairy consumption during pregnancy influence fetal growth and infant birthweight? A systematic literature review. Food Nutr Res. 2012;56:20050. Brantsæter AL, Olafsdottir AS, Forsum E, Olsen SF, Thorsdottir I. Does milk and dairy consumption during pregnancy influence fetal growth and infant birthweight? A systematic literature review. Food Nutr Res. 2012;56:20050.
163.
Zurück zum Zitat Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290–6.PubMed Boney CM, Verma A, Tucker R, Vohr BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005;115:e290–6.PubMed
164.
Zurück zum Zitat Sørensen HT, Sabroe S, Rothman KJ, Gillman M, Fischer P, Sørensen TI. Relation between weight and length at birth and body mass index in young adulthood: cohort study. BMJ. 1997;315:1137.PubMedCentralPubMed Sørensen HT, Sabroe S, Rothman KJ, Gillman M, Fischer P, Sørensen TI. Relation between weight and length at birth and body mass index in young adulthood: cohort study. BMJ. 1997;315:1137.PubMedCentralPubMed
165.
Zurück zum Zitat Leunissen RW, Stijnen T, Hokken-Koelega AC. Influence of birth size on body composition in early adulthood: the programming factors for growth and metabolism (PROGRAM)-study. Clin Endocrinol (Oxf). 2009;70:245–51. Leunissen RW, Stijnen T, Hokken-Koelega AC. Influence of birth size on body composition in early adulthood: the programming factors for growth and metabolism (PROGRAM)-study. Clin Endocrinol (Oxf). 2009;70:245–51.
166.
Zurück zum Zitat Brüske I, Flexeder C, Heinrich J. Body mass index and incidence of asthma in children. Curr Opin Allergy Clin Immunol. 2014;14:155–60.PubMed Brüske I, Flexeder C, Heinrich J. Body mass index and incidence of asthma in children. Curr Opin Allergy Clin Immunol. 2014;14:155–60.PubMed
167.
Zurück zum Zitat Skilton MR, Siitonen N, Würtz P, Viikari JS, Juonala M, Seppälä I, et al. High birth weight is associated with obesity and increased carotid wall thickness in young adults: the cardiovascular risk in young Finns study. Arterioscler Thromb Vasc Biol. 2014;34:1064–8.PubMed Skilton MR, Siitonen N, Würtz P, Viikari JS, Juonala M, Seppälä I, et al. High birth weight is associated with obesity and increased carotid wall thickness in young adults: the cardiovascular risk in young Finns study. Arterioscler Thromb Vasc Biol. 2014;34:1064–8.PubMed
168.
Zurück zum Zitat Bukowski R, Chlebowski RT, Thune I, Furberg AS, Hankins GD, Malone FD, et al. Birth weight, breast cancer and the potential mediating hormonal environment. PLoS One. 2012;7:e40199.PubMedCentralPubMed Bukowski R, Chlebowski RT, Thune I, Furberg AS, Hankins GD, Malone FD, et al. Birth weight, breast cancer and the potential mediating hormonal environment. PLoS One. 2012;7:e40199.PubMedCentralPubMed
169.
Zurück zum Zitat Spracklen CN, Wallace RB, Sealy-Jefferson S, Robinson JG, Freudenheim JL, Wellons MF, et al. Birth weight and subsequent risk of cancer. Cancer Epidemiol. 2014;38:538–43.PubMed Spracklen CN, Wallace RB, Sealy-Jefferson S, Robinson JG, Freudenheim JL, Wellons MF, et al. Birth weight and subsequent risk of cancer. Cancer Epidemiol. 2014;38:538–43.PubMed
170.
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.PubMed 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.PubMed
171.
Zurück zum Zitat Yang Z, Huffman SL. Nutrition in pregnancy and early childhood and associations with obesity in developing countries. Matern Child Nutr. 2013;9 Suppl 1:105–19.PubMed Yang Z, Huffman SL. Nutrition in pregnancy and early childhood and associations with obesity in developing countries. Matern Child Nutr. 2013;9 Suppl 1:105–19.PubMed
172.
Zurück zum Zitat Haissaguerre M, Saucisse N, Cota D. Influence of mTOR in energy and metabolic homeostasis. Mol Cell Endocrinol. 2014;397:67–77.PubMed Haissaguerre M, Saucisse N, Cota D. Influence of mTOR in energy and metabolic homeostasis. Mol Cell Endocrinol. 2014;397:67–77.PubMed
173.
Zurück zum Zitat Melnik BC. The potential mechanistic link between allergy and obesity development and infant formula feeding. Allergy Asthma Clin Immunol. 2014;10:37.PubMedCentralPubMed Melnik BC. The potential mechanistic link between allergy and obesity development and infant formula feeding. Allergy Asthma Clin Immunol. 2014;10:37.PubMedCentralPubMed
174.
Zurück zum Zitat Melnik BC. Excessive leucine-mTORC1-signalling of cow milk-based infant formula: The missing link to understand early childhood obesity. J Obes. 2012;2012:197653.PubMedCentralPubMed Melnik BC. Excessive leucine-mTORC1-signalling of cow milk-based infant formula: The missing link to understand early childhood obesity. J Obes. 2012;2012:197653.PubMedCentralPubMed
175.
Zurück zum Zitat Harlan SM, Guo DF, Morgan DA, Fernandes-Santos C, Rahmouni K. Hypothalamic mTORC1 signaling controls sympathetic nerve activity and arterial pressure and mediates leptin effects. Cell Metab. 2013;17:599–606.PubMedCentralPubMed Harlan SM, Guo DF, Morgan DA, Fernandes-Santos C, Rahmouni K. Hypothalamic mTORC1 signaling controls sympathetic nerve activity and arterial pressure and mediates leptin effects. Cell Metab. 2013;17:599–606.PubMedCentralPubMed
176.
Zurück zum Zitat Villanueva EC, Münzberg H, Cota D, Leshan RL, Kopp K, Ishida-Takahashi R, et al. Complex regulation of mammalian target of rapamycin complex 1 in the basomedial hypothalamus by leptin and nutritional status. Endocrinology. 2009;150:4541–51.PubMedCentralPubMed Villanueva EC, Münzberg H, Cota D, Leshan RL, Kopp K, Ishida-Takahashi R, et al. Complex regulation of mammalian target of rapamycin complex 1 in the basomedial hypothalamus by leptin and nutritional status. Endocrinology. 2009;150:4541–51.PubMedCentralPubMed
177.
Zurück zum Zitat Oken E, Gillman MW. Fetal origins of obesity. Obes Res. 2003;11:496–506.PubMed Oken E, Gillman MW. Fetal origins of obesity. Obes Res. 2003;11:496–506.PubMed
178.
Zurück zum Zitat Gillman MW. A life course approach to obesity. In: Kuh D, Be-Shlomo Y, editors. A life course approach to chronic disease epidemiology. 2nd ed. New York, NY: Oxford University Press; 2004. p. 189–217. Gillman MW. A life course approach to obesity. In: Kuh D, Be-Shlomo Y, editors. A life course approach to chronic disease epidemiology. 2nd ed. New York, NY: Oxford University Press; 2004. p. 189–217.
179.
Zurück zum Zitat Freinkel N. Banting Lecture 1980. Of pregnancy and progeny. Diabetes. 1980;29:1023–35.PubMed Freinkel N. Banting Lecture 1980. Of pregnancy and progeny. Diabetes. 1980;29:1023–35.PubMed
180.
Zurück zum Zitat Lynch CJ, Adams SH. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 2014;10:723–36.PubMed Lynch CJ, Adams SH. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 2014;10:723–36.PubMed
181.
Zurück zum Zitat Whitaker RC, Dietz WH. Role of the prenatal environment in the development of obesity. J Pediatr. 1998;132:768–76.PubMed Whitaker RC, Dietz WH. Role of the prenatal environment in the development of obesity. J Pediatr. 1998;132:768–76.PubMed
182.
Zurück zum Zitat Melnik BC. Formula feeding promotes adipogenic, diabetogenic, hypertonic and allergic mTORC1-programming. In: Preedy VR, Watson RR, Zibadi S, editors. Handbook of dietary and nutritional aspects of bottle feeding. Human Health Handbooks no. 8, Wageningen Academic Publishers Wageningen. 2014. p. 545–68. Melnik BC. Formula feeding promotes adipogenic, diabetogenic, hypertonic and allergic mTORC1-programming. In: Preedy VR, Watson RR, Zibadi S, editors. Handbook of dietary and nutritional aspects of bottle feeding. Human Health Handbooks no. 8, Wageningen Academic Publishers Wageningen. 2014. p. 545–68.
184.
Zurück zum Zitat Melnik BC, John SM, Schmitz G. Milk: an exosomal microRNA transmitter promoting thymic regulatory T cell maturation preventing the development of atopy? J Transl Med. 2014;12:43.PubMedCentralPubMed Melnik BC, John SM, Schmitz G. Milk: an exosomal microRNA transmitter promoting thymic regulatory T cell maturation preventing the development of atopy? J Transl Med. 2014;12:43.PubMedCentralPubMed
185.
Zurück zum Zitat Dodd JM, McPhee AJ, Turnbull D, Yelland LN, Deussen AR, Grivell RM, et al. The effects of antenatal dietary and lifestyle advice for women who are overweight or obese on neonatal health outcomes: the LIMIT randomized trial. BMC Med. 2014;12:163.PubMedCentralPubMed Dodd JM, McPhee AJ, Turnbull D, Yelland LN, Deussen AR, Grivell RM, et al. The effects of antenatal dietary and lifestyle advice for women who are overweight or obese on neonatal health outcomes: the LIMIT randomized trial. BMC Med. 2014;12:163.PubMedCentralPubMed
Metadaten
Titel
Milk consumption during pregnancy increases birth weight, a risk factor for the development of diseases of civilization
verfasst von
Bodo C Melnik
Swen Malte John
Gerd Schmitz
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2015
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-014-0377-9

Weitere Artikel der Ausgabe 1/2015

Journal of Translational Medicine 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.