Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Life Sci ; 337: 122344, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38081408

RESUMEN

BACKGROUND: Maternal obesity (MO) increases fetal androgen concentrations, the prevalence of macrosomia, and predisposes offspring to metabolic dysfunction in later life, especially males. These risks may be, in part, the result of increased liver-specific androgen signalling pathway activity in utero. Androgen signalling activity can be suppressed by androgen metabolism via cytochrome P450 (CYP) isoenzymes (CYP2B6, CYP3A) or through inhibition of the full-length androgen receptor (AR-FL) via the antagonistic isoform, AR-45. We hypothesised MO impairs CYP enzyme activity and AR-45 expression in male fetal livers, thereby enhancing activity of androgen signalling pathways. METHODS: Nine months prior to pregnancy, nulliparous female baboons were assigned to either ad libitum control or high fat diet. At 165 day (d) gestation (term, 180 d) fetal liver was collected (n = 6/sex/group). CYP activity was quantified using functional assays; subcellular AR expression was measured using Western blot. RESULTS: CYP2B6 and CYP3A activity, and nuclear expression of AR-45, was reduced in MO males only. Nuclear AR-45 expression was inversely related with fetal body weight of MO males only. CONCLUSIONS: Reduced CYP2B6 and CYP3A activity in conjunction with decreased nuclear AR-45 expression may enhance liver androgen signalling in males from MO pregnancies, thereby increasing the risk of macrosomia, as well as metabolic dysfunction in later life.


Asunto(s)
Andrógenos , Obesidad Materna , Humanos , Femenino , Embarazo , Masculino , Andrógenos/metabolismo , Obesidad Materna/metabolismo , Citocromo P-450 CYP3A/metabolismo , Citocromo P-450 CYP2B6/metabolismo , Macrosomía Fetal/metabolismo , Receptores Androgénicos/metabolismo , Hígado/metabolismo , Isoenzimas
2.
Commun Biol ; 6(1): 1226, 2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-38049528

RESUMEN

The maternal liver is challenged by metabolic demands throughout pregnancy. However, hepatocyte dynamics and their physiological significance in pregnancy remain unclear. Here, we show in mice that hepatocyte proliferation is spatiotemporally regulated in each liver lobular zone during pregnancy, with transient proliferation of periportal and pericentral hepatocytes during mid and late gestation, respectively. Using adeno-associated virus (AAV)-8-mediated expression of the cell cycle inhibitor p21 in hepatocytes, we show that inhibition of hepatocyte proliferation during mid, but not late, gestation impairs liver growth. Transcriptionally, genes involved in glucose/glycogen metabolism are downregulated in late pregnancy when midgestational hepatocyte proliferation is attenuated. In addition, hepatic glycogen storage is abolished, with concomitant elevated blood glucose concentrations, glucose intolerance, placental glycogen deposition, and fetal overgrowth. Laser capture microdissection and RNA-seq analysis of each liver lobular zone show zone-specific changes in the transcriptome during pregnancy and identify genes that are periportally expressed at midgestation, including the hyaluronan-mediated motility receptor (Hmmr). Knockdown of Hmmr in hepatocytes by AAV8-shHmmr suppresses periportal hepatocyte proliferation at midgestation and induces impaired hepatic glycogen storage, glucose intolerance, placental glycogen deposition and fetal overgrowth. Our results suggest that periportal hepatocyte proliferation during midgestation is critical for maternal glycogen metabolism and fetal size.


Asunto(s)
Diabetes Gestacional , Intolerancia a la Glucosa , Humanos , Ratones , Embarazo , Femenino , Animales , Glucógeno Hepático/metabolismo , Placenta/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Macrosomía Fetal/metabolismo , Glucosa/metabolismo , Glucógeno/metabolismo , Hepatocitos/metabolismo , Homeostasis , Proliferación Celular
3.
Clin Sci (Lond) ; 137(21): 1651-1664, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37861075

RESUMEN

The System L amino acid transporter, particularly the isoform Large Neutral Amino Acid Transporter Small Subunit 1 (LAT1) encoded by SLC7A5, is believed to mediate the transfer of essential amino acids in the human placenta. Placental System L amino acid transporter expression and activity is decreased in pregnancies complicated by IUGR and increased in fetal overgrowth. However, it remains unknown if changes in the expression of LAT1 are mechanistically linked to System L amino acid transport activity. Here, we combined overexpression approaches with protein analysis and functional studies in cultured primary human trophoblast (PHT) cells to test the hypothesis that SLC7A5 overexpression increases the uptake of essential amino acids and activates mTOR signaling in PHT cells. Overexpression of SLC7A5 resulted in a marked increase in protein expression of LAT1 in the PHT cells microvillous plasma membrane and System L amino acid transporter activity. Moreover, mTOR signaling was activated, and System A amino acid transporter activity increased following SLC7A5 overexpression, suggesting coordination of trophoblast amino transporter expression and activity to ensure balanced nutrient flux to the fetus. This is the first report showing that overexpression of LAT1 is sufficient to increase the uptake of essential amino acids in PHT cells, which activates mTOR, a master regulator of placental function. The decreased placental System L activity in human IUGR and the increased placental activity of this transporter system in some cases of fetal overgrowth may directly contribute to changes in fetal amino acid availability and altered fetal growth in these pregnancy complications.


Asunto(s)
Diabetes Gestacional , Trofoblastos , Femenino , Humanos , Embarazo , Aminoácidos/metabolismo , Aminoácidos Esenciales/metabolismo , Diabetes Gestacional/metabolismo , Macrosomía Fetal/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Placenta/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Trofoblastos/metabolismo
4.
J Mol Endocrinol ; 71(4)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37855320

RESUMEN

Suboptimal in utero environments such as poor maternal nutrition and gestational diabetes can impact fetal birth weight and the metabolic health trajectory of the adult offspring. Fetal growth is associated with alterations in placental mechanistic target of rapamycin (mTOR) signaling; it is reduced in fetal growth restriction and increased in fetal overgrowth. We previously reported that when metabolically challenged by a high-fat diet, placental mTORKO (mTORKOpl) adult female offspring develop obesity and insulin resistance, whereas placental TSC2KO (TSC2KOpl) female offspring are protected from diet-induced obesity and maintain proper glucose homeostasis. In the present study, we sought to investigate whether reducing or increasing placental mTOR signaling in utero alters the programming of adult offspring metabolic tissues preceding a metabolic challenge. Adult male and female mTORKOpl, TSC2KOpl, and respective controls on a normal chow diet were subjected to an acute intraperitoneal insulin injection. Upon insulin stimulation, insulin signaling via phosphorylation of Akt and nutrient sensing via phosphorylation of mTOR target ribosomal S6 were evaluated in the offspring liver, white adipose tissue, and skeletal muscle. Among tested tissues, we observed significant changes only in the liver signaling. In the male mTORKOpl adult offspring liver, insulin-stimulated phospho-Akt was enhanced compared to littermate controls. Basal phospho-S6 level was increased in the mTORKOpl female offspring liver compared to littermate controls and did not increase further in response to insulin. RNA sequencing of offspring liver identified placental mTORC1 programming-mediated differentially expressed genes. The expression of major urinary protein 1 (Mup1) was differentially altered in female mTORKOpl and TSC2KOpl offspring livers and we show that MUP1 level is dependent on overnutrition and fasting status. In summary, deletion of placental mTOR nutrient sensing in utero programs hepatic response to insulin action in a sexually dimorphic manner. Additionally, we highlight a possible role for hepatic and circulating MUP1 in glucose homeostasis that warrants further investigation.


Asunto(s)
Diabetes Gestacional , Placenta , Animales , Femenino , Masculino , Ratones , Embarazo , Diabetes Gestacional/metabolismo , Macrosomía Fetal/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Placenta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
5.
Clin Sci (Lond) ; 137(8): 663-678, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37014924

RESUMEN

Pregnant women with obesity are more likely to deliver infants who are large for gestational age (LGA). LGA is associated with increased perinatal morbidity and risk of developing metabolic disease later in life. However, the mechanisms underpinning fetal overgrowth remain to be fully established. Here, we identified maternal, placental, and fetal factors that are associated with fetal overgrowth in pregnant women with obesity. Maternal and umbilical cord plasma and placentas were collected from women with obesity delivering infants who were LGA (n=30) or appropriate for gestational age (AGA, n=21) at term. Maternal and umbilical cord plasma analytes were measured using multiplex sandwich assay and ELISA. Insulin/mechanistic target of rapamycin (mTOR) signaling activity was determined in placental homogenates. Amino acid transporter activity was measured in isolated syncytiotrophoblast microvillous membrane (MVM) and basal membrane (BM). Glucagon-like peptide-1 receptor (GLP-1R) protein expression and signaling were measured in cultured primary human trophoblast (PHT) cells. Maternal plasma glucagon-like peptide-1 (GLP-1) was higher in LGA pregnancies and positively correlated to birthweight. Umbilical cord plasma insulin, C-peptide, and GLP-1 were increased in obese-large for gestational age (OB-LGA) infants. LGA placentas were larger but showed no change in insulin/mTOR signaling or amino acid transport activity. GLP-1R protein was expressed in the MVM isolated from human placenta. GLP-1R activation stimulated protein kinase alpha (PKA), extracellular signal-regulated kinase-1 and-2 (ERK1/2), and mTOR pathways in PHT cells. Our results suggest elevated maternal GLP-1 may drive fetal overgrowth in obese pregnant women. We speculate that maternal GLP-1 acts as a novel regulator of fetal growth by promoting placental growth and function.


Asunto(s)
Diabetes Gestacional , Placenta , Femenino , Humanos , Embarazo , Diabetes Gestacional/metabolismo , Desarrollo Fetal , Macrosomía Fetal/complicaciones , Macrosomía Fetal/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Placenta/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Péptido 1 Similar al Glucagón
6.
Pediatr Res ; 93(1): 267-273, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35459765

RESUMEN

BACKGROUND: Peroxisomal proliferator-activated receptors (PPARs) and microRNAs (miRNAs) play important roles in the development of fetuses, whereas expression changes of PPARs and three miRNAs (miR-17, miR-27b and miR-34a) and whether these miRNAs regulate PPARs in non-GDM macrosomia placenta is unclear. METHODS: A case-control study was performed to collect information and placental tissues on mothers and newborns of non-GDM macrosomia and normal-birth-weight infants. In vitro HTR8-SVneo cellular model was used to detect the effects of miRNAs on PPARs expression. Quantitative real-time PCR (qRT-PCR) and western blot was applied to examine the expression levels of PPARs, miR-17, miR-27b, and miR-34a in placental tissues and cells. RESULTS: The PPARα/γ mRNA and protein levels were significantly up-regulated and miR-27b was down-regulated in the placenta of macrosomia group compared with in the control group, while no difference was observed in PPARß, miR-17, and miR-34a. After adjusting for confounding factors, low miR-27b and high PPARα/γ mRNA expression still increased the risk of macrosomia. The PPARα/γ protein levels presented a corresponding decrease or increase when cells were transfected with miR-27b mimic or inhibitor. CONCLUSIONS: Placental PPARα/γ and miR-27b expression were associated with non-GDM macrosomia and miR-27b probably promotes the occurrence of non-GDM macrosomia by regulating PPARα/γ protein. IMPACT: Low miR-27b and high PPARα/γ mRNA expression in the placenta were associated with higher risk of macrosomia. In vitro HTR8-SVneo cell experiment supported that miR-27b could negatively regulate the expression of PPARα and PPARγ protein. MiR-27b was probably involved in non-GDM macrosomia through negative regulation of PPARα/γ protein.


Asunto(s)
MicroARNs , Placenta , Recién Nacido , Humanos , Embarazo , Femenino , Placenta/metabolismo , Macrosomía Fetal/genética , Macrosomía Fetal/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Estudios de Casos y Controles , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/metabolismo
7.
Clin Epigenetics ; 14(1): 192, 2022 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-36585686

RESUMEN

BACKGROUND: Fetal overgrowth "programs" an elevated risk of type 2 diabetes in adulthood. Epigenetic alterations may be a mechanism in programming the vulnerability. We sought to characterize genome-wide alterations in placental gene methylations in fetal overgrowth and the associations with metabolic health biomarkers including leptin, adiponectin and fetal growth factors. RESULTS: Comparing genome-wide placental gene DNA methylations in large-for-gestational-age (LGA, an indicator of fetal overgrowth, n = 30) versus optimal-for-gestational-age (OGA, control, n = 30) infants using the Illumina Infinium Human Methylation-EPIC BeadChip, we identified 543 differential methylation positions (DMPs; 397 hypermethylated, 146 hypomethylated) at false discovery rate < 5% and absolute methylation difference > 0.05 after adjusting for placental cell-type heterogeneity, maternal age, pre-pregnancy BMI and HbA1c levels during pregnancy. Twenty-five DMPs annotated to 20 genes (QSOX1, FCHSD2, LOC101928162, ADGRB3, GCNT1, TAP1, MYO16, NAV1, ATP8A2, LBXCOR1, EN2, INCA1, CAMTA2, SORCS2, SLC4A4, RPA3, UMAD1,USP53, OR2L13 and NR3C2) could explain 80% of the birth weight variations. Pathway analyses did not detect any statistically significant pathways after correcting for multiple tests. We validated a newly discovered differentially (hyper-)methylated gene-visual system homeobox 1 (VSX1) in an independent pyrosequencing study sample (LGA 47, OGA 47). Our data confirmed a hypermethylated gene-cadherin 13 (CDH13) reported in a previous epigenome-wide association study. Adiponectin in cord blood was correlated with its gene methylation in the placenta, while leptin and fetal growth factors (insulin, IGF-1, IGF-2) were not. CONCLUSIONS: Fetal overgrowth may be associated with a large number of altered placental gene methylations. Placental VSX1 and CDH13 genes are hypermethylated in fetal overgrowth. Placental ADIPOQ gene methylations and fetal circulating adiponectin levels were correlated, suggesting the contribution of placenta-originated adiponectin to cord blood adiponectin.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Embarazo , Femenino , Humanos , Adulto , Placenta/metabolismo , Metilación de ADN , Leptina/genética , Adiponectina , Diabetes Gestacional/genética , Diabetes Mellitus Tipo 2/genética , Macrosomía Fetal/genética , Macrosomía Fetal/metabolismo , Edad Gestacional , Sangre Fetal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Desarrollo Fetal/genética , Proteínas Portadoras/genética , Proteínas de la Membrana/genética
8.
J Endocrinol ; 254(1): 37-49, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35674008

RESUMEN

The aim of this study was to evaluate the paternal programming of sex-dependent alterations in fetoplacental growth and placental lipid metabolism regulated by peroxisome proliferator-activated receptor (PPAR) target genes in F1 diabetic males born from F0 pregestational diabetic rats. F1 control and diabetic male rats were mated with control female rats. On day 21 of gestation, F2 male and female fetoplacental growth, placental lipid levels, and protein and mRNA levels of genes involved in lipid metabolism and transport were evaluated. Fetal but not placental weight was increased in the diabetic group. Triglyceride, cholesterol and free fatty acid levels were increased in placentas of male fetuses from the diabetic group. The mRNA levels of Pparα and Pparγ coactivator 1α (Pgc-1α) were increased only in placentas of male fetuses from the diabetic group. Protein levels of PPARα and PGC-1α were decreased only in placentas of male fetuses from the diabetic group. No differences were found in Pparγ mRNA and protein levels in placentas from the diabetic group. The mRNA levels of genes involved in lipid synthesis showed no differences between groups, whereas the mRNA levels of genes involved in lipid oxidation and transport were increased only in placentas of male fetuses from the diabetic group. In conclusion, paternal diabetes programs fetal overgrowth and sex-dependent effects on the regulation of lipid metabolism in the placenta, where only placentas of male fetuses show an increase in lipid accumulation and mRNA expression of enzymes involved in lipid oxidation and transport pathways.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Gestacional , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Femenino , Macrosomía Fetal/metabolismo , Humanos , Masculino , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Placenta/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Triglicéridos/metabolismo
9.
Mol Med Rep ; 26(2)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35674181

RESUMEN

Macrosomia is a common perinatal complication, with a series of adverse effects on newborns and pregnant women. However, the effects of long non­coding RNAs (lncRNAs) on non­diabetic fetal macrosomia (NDFMS) remain unclear. The aim of the present study was to investigate whether aberrant lncRNA expression in the placenta is involved in the pathogenesis of NDFMS and to elucidate its biological mechanisms. The expression profile of lncRNAs in the placentas of pregnant women with NDFMS was investigated using an Agilent Human LncRNA Microarray. Differentially expressed lncRNAs were selected for validation using reverse transcription­quantitative polymerase chain reaction (RT­qPCR). Additionally, the function of lncRNA ubiquitin­specific peptidase 2 antisense RNA 1 (USP2­AS1) was investigated using a trophoblast cell line. The results revealed that 763 lncRNAs were upregulated and 129 lncRNAs were downregulated in the placentas of women in the NDFMS group (|FC| ≥2.0). A total of 10 lncRNAs (|FC| ≥4.0, signal value ≥50) were selected for validation using two­stage RT­qPCR, indicating that the expression trends of the 10 differentially expressed lncRNAs in the NDFMS group (n=8 vs. 8 and 48 vs. 48) were consistent with the microarray data. In addition, a significant downregulation in the levels of lncRNA USP2­AS1 was observed in both the microarray data and second­stage verification. The overexpression of lncRNA USP2­AS1 induced G1 phase cell cycle arrest and the number of cells entering S phase was reduced. In addition, the viability of HTR­8/SVneo cells was significantly inhibited when lncRNA USP2­AS1 was overexpressed. Therefore, these findings demonstrated that lncRNAs were significantly differentially expressed in the placentas of pregnant women with NDFMS and that the downregulation of lncRNA USP2­AS1 may be involved in the pathogenesis of NDFMS, by promoting trophoblast cell viability.


Asunto(s)
ARN Largo no Codificante , Proliferación Celular , Regulación hacia Abajo , Femenino , Macrosomía Fetal/genética , Macrosomía Fetal/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Recién Nacido , Placenta/metabolismo , Embarazo , Mujeres Embarazadas , ARN sin Sentido/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Trofoblastos/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
10.
FASEB J ; 36(7): e22383, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35670755

RESUMEN

Infants born to obese mothers have a greater risk for childhood obesity and insulin resistance. However, the underlying biological mechanism remains elusive, which constitutes a significant roadblock for developing specific prevention strategies. Maternal adiponectin levels are lower in obese pregnant women, which is linked with increased placental nutrient transport and fetal overgrowth. We have previously reported that adiponectin supplementation to obese dams during the last four days of pregnancy prevented the development of obesity, glucose intolerance, muscle insulin resistance, and fatty liver in three months old offspring. In the present study, we tested the hypothesis that 6-9-month-old offspring of obese dams show glucose intolerance associated with muscle insulin resistance and mitochondrial dysfunction and that normalization of maternal adiponectin in obese pregnant mice prevents the development of this phenotype in the offspring. Male and female offspring of obese mice exhibited in vivo glucose intolerance and insulin resistance at 6 and 9 months of age. In gastrocnemius muscles ex vivo, male and female offspring of obese dams showed reduced phosphorylation of insulin receptor substrate 1Tyr-608 , AktThr-308 , and decreased Glut4 plasma membrane translocation upon insulin stimulation. These metabolic abnormalities in offspring born to obese mice were largely prevented by normalization of maternal adiponectin levels in late pregnancy. We provide evidence that low circulating maternal adiponectin is a critical mechanistic link between maternal obesity and the development of metabolic disease in offspring. Strategies aimed at improving maternal adiponectin levels may prevent long-term metabolic dysfunction in offspring of obese mothers.


Asunto(s)
Diabetes Gestacional , Intolerancia a la Glucosa , Resistencia a la Insulina , Adiponectina/metabolismo , Animales , Diabetes Gestacional/metabolismo , Femenino , Macrosomía Fetal/metabolismo , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/prevención & control , Insulina/metabolismo , Masculino , Ratones , Ratones Obesos , Placenta/metabolismo , Embarazo
11.
J Perinat Med ; 50(7): 947-960, 2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-35363447

RESUMEN

OBJECTIVES: Diabetes mellitus (DM) in pregnancy and gestational diabetes remain a considerable cause of pregnancy complications, and fetal macrosomia is among them. Insulin, insulin-like growth factors (IGFs), and components of their signal-transduction axes belong to the predominant growth regulators and are implicated in glucose homeostasis. This study aimed to evaluate the available evidence on the association between the IGF axis and fetal anthropometric parameters in human diabetic pregnancy. METHODS: PubMed, Medline, Web of Science, and CNKI databases (1981-2021) were searched. RESULTS: Maternal and cord serum IGF-I levels are suggested to be positively associated with weight and length of neonates born to mothers with type 1 DM. The results concerning IGF-II and IGFBPs in type 1 DM or any of the IGF axis components in type 2 DM remain controversial. The alterations of maternal serum IGFs concentrations throughout diabetic and non-diabetic pregnancy do not appear to be the same. Maternal 1st trimester IGF-I level is positively associated with fetal birth weight in DM. CONCLUSIONS: Research on the IGF axis should take gestational age of sampling, presence of DM, and insulin administration into account. Maternal 1st trimester IGF-I level might become a predictor for macrosomia development in diabetic pregnancy.


Asunto(s)
Diabetes Mellitus Tipo 1 , Embarazo en Diabéticas , Peso al Nacer , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Sangre Fetal/metabolismo , Macrosomía Fetal/etiología , Macrosomía Fetal/metabolismo , Edad Gestacional , Humanos , Recién Nacido , Insulina , Factor I del Crecimiento Similar a la Insulina/metabolismo , Embarazo , Embarazo en Diabéticas/metabolismo
12.
PLoS One ; 17(4): e0265419, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35390031

RESUMEN

Gestational diabetes mellitus (GDM) is one of the most common complications of pregnancy but the underlying mechanism remains obscure. The aims of this study are to examine if omental adipose tissue (OMAT) and subcutaneous AT (SCAT) differentially express proinflammatory and lipid metabolic adipokines, and if so, whether their regional differences have implications on lipid metabolism in GDM. Paired samples of OMAT and SCAT were excised from pregnant women in scheduled Cesarean sections with non-obese (NOBS), obese (OBS) and GDM. The results showed that the mRNA of monocyte chemoattractant protein (MCP)-1, macrophage marker CD68, and cytokines IL-6, IL-8, and TNF-α are increased in OMAT from GDM women compared to that in NOBS and OBS women (P<0.05). Glucose and TNF-α dose-dependently enhanced ADM and its receptor components CRLR and RAMPs in human adipocytes. Immunofluorescence showed that ADM and its receptor components are higher in OMAT from GDM women compared to non-GDM women. Further, basal lipolysis was greater in OMAT than in SCAT and ADM stimulates further glycerol release in OMAT, but not in SCAT, and these increases are reduced by ADM antagonist, ADM22-52. We therefore conclude that elevated ADM and its receptor expressions by OMAT, but not by SCAT appear to contribute to the lipid dysregulation in GDM women, and manipulation of ADM may represent one of the novel approaches in minimizing the risk of GDM-related fetal overgrowth.


Asunto(s)
Tejido Adiposo , Adrenomedulina , Diabetes Gestacional , Grasa Subcutánea , Tejido Adiposo/metabolismo , Adrenomedulina/metabolismo , Diabetes Gestacional/metabolismo , Femenino , Macrosomía Fetal/metabolismo , Humanos , Lípidos , Obesidad/genética , Obesidad/metabolismo , Epiplón , Embarazo , Grasa Subcutánea/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Reprod Sci ; 29(3): 904-914, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34750770

RESUMEN

To assess associations between infants with macrosomia and placental expression levels of lipid activated/transport-related factors and umbilical cord blood lipid concentrations in healthy pregnancy. We conducted a case-control study of 38 macrosomic neonates (MS group) and 39 normal-birth-weight newborns (NC group) in a healthy pregnancy. Cord blood lipid levels were measured by automatic biochemical analyzer, mRNA and protein expression levels of placental lipid activated/transport-related factors were determined by real-time polymerase chain reaction and western blot, respectively. Compared with NC group, cord blood total cholesterol (TC), low-density lipoprotein cholesterol (LDLC), and non-esterified fatty acid (NEFA) concentrations were decreased in the MS group. The mRNA and protein expression levels of placental peroxisome proliferator-activated receptors (PPARα, PPARγ), plasma membrane fatty acid-binding protein (FABPpm), and fatty acid translocase (FAT/CD36) were significantly higher in the MS group than the NC group. And there was a weak positive correlation between the expression of PPARγ, FABP4, and FABP3 mRNA in the placenta and the HDLC (rs = 0.439; P = 0.005), NEFA (rs = 0.342; P = 0.041), and TG (rs = 0.349; P = 0.034) levels in the cord blood in the MS group, respectively. After multivariate adjustment, the logistic regression analysis showed that high placental PPARα (adjusted odds ratio [AOR] = 3.022; 95% confidence interval [CI] 1.032-8.853) and FAT/CD36 (AOR=2.989; 95%CI 1.029-8.679) and low LDLC concentration in the cord blood (AOR=0.246; 95%CI 0.080-0.759) increased the risk of macrosomia. The increased PPARα and FAT/CD36 expression levels may influence the occurrence of fetal macrosomia through regulating placental lipid transport.


Asunto(s)
Proteínas de Transporte de Ácidos Grasos/sangre , Macrosomía Fetal/metabolismo , Placenta/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Sangre Fetal/metabolismo , Humanos , Recién Nacido , Embarazo
14.
J Clin Endocrinol Metab ; 107(2): e688-e697, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34477199

RESUMEN

CONTEXT: Fetal overgrowth "programs" an elevated risk of obesity and type 2 diabetes in adulthood. Plausibly, adipokines may be involved in programming metabolic health. OBJECTIVE: This work aimed to evaluate whether large-for-gestational-age (LGA), an indicator of fetal overgrowth, is associated with altered circulating leptin and adiponectin levels in infancy, and assess the determinants. METHODS: In the Canadian 3D birth cohort, we studied 70 LGA (birth weight > 90th percentile) and 140 optimal-for-gestational-age (OGA, 25th-75th percentiles) infants matched by maternal ethnicity, smoking, and gestational age at delivery. The primary outcomes were fasting leptin, and total and high-molecular-weight (HMW) adiponectin concentrations at age 2 years. RESULTS: LGA infants had higher body mass index (BMI) than OGA infants. However, there were no significant differences in leptin, and total and HMW adiponectin concentrations. Leptin concentrations were positively associated with female sex, weight (z score) gain 0 to 24 months, current BMI, and the sum of triceps and subscapular skinfold thickness, and negatively associated with maternal age and White ethnicity. Female sex was associated with lower total and HMW adiponectin concentrations. Weight (z score) gain 0 to 24 months and current BMI were positively correlated with total and HMW adiponectin concentrations in LGA infants only. CONCLUSION: This study is the first to demonstrate that LGA does not matter for circulating leptin and adiponectin concentrations in infancy, and there may be LGA-specific positive associations between weight gain or current BMI and adiponectin concentrations in infancy, suggesting dysfunction in establishing the adiposity-adiponectin negative feedback loop in LGA individuals.


Asunto(s)
Adiponectina/sangre , Macrosomía Fetal/metabolismo , Resistencia a la Insulina , Leptina/sangre , Aumento de Peso , Adiponectina/metabolismo , Adiposidad/fisiología , Peso al Nacer/fisiología , Canadá , Estudios de Casos y Controles , Preescolar , Femenino , Macrosomía Fetal/sangre , Macrosomía Fetal/complicaciones , Estudios de Seguimiento , Edad Gestacional , Humanos , Lactante , Recién Nacido , Leptina/metabolismo , Masculino , Factores Sexuales
15.
Reprod Sci ; 29(2): 596-605, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34270000

RESUMEN

Gestational diabetes mellitus (GDM) has become a worldwide problem in recent years. Macrosomia, a primary consequence of GDM, has short-term and life-long consequences in the offspring of mothers with GDM. Our previous study showed that miR-517a was dysregulated in placenta and plasma of fetal growth restriction through inhibiting invasion of trophoblast and might be closely related with the regulation of birth weight by the placenta. To further investigate the mechanism of miR-517a, we conducted genome-wide microarray profile of lncRNAs. lncRNA-SNX17 was found to be significantly upregulated in the placenta of diabetic macrosomia by qRT-PCR, and the expression of miR-517a and IGF-1 were measured by qRT-PCR and Western blot. Interestingly, significant inverse correlations of the miR-517a with both lncRNA-SNX17 and IGF-1 expression were revealed in the placenta of diabetic macrosomia. Bioinformatic prediction also revealed that both lncRNA-SNX17 and IGF-1 possessed binding sites for miR-517a, which were then confirmed by luciferase report assay. LncRNA-SNX17 overexpression reduced the expression of miR-517a and increased the IGF-1 expression in HTR-8/SVneo human trophoblast cell line and thus enhanced the proliferation of HTR-8/SVneo. The enhancement of HTR-8/SVneo proliferation by lncRNA-SXN17 could be nullified by co-transfection of miR-517a mimics. The data suggested that lncRNA-SNX17 might promote the trophoblast proliferation through miR-517a/IGF-1 pathway and might play a role in the placentation of diabetic macrosomia.


Asunto(s)
Diabetes Gestacional/metabolismo , Macrosomía Fetal/etiología , Macrosomía Fetal/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/metabolismo , Placenta/metabolismo , Embarazo en Diabéticas/metabolismo , ARN Largo no Codificante/metabolismo , Adulto , Western Blotting , Línea Celular , Femenino , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Trofoblastos/metabolismo
16.
Reprod Toxicol ; 104: 16-26, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34175429

RESUMEN

Experimental models of maternal diabetes lead to the intrauterine programming of Gestational Diabetes Mellitus (GDM) in the offspring, together with an intrauterine proinflammatory environment, feto-placental metabolic alterations and fetal overgrowth. The aim of this work was to evaluate the effect of the mitochondrial antioxidant Idebenone given to F0 mild pregestational diabetic rats on the development of GDM in their F1 offspring and the intergenerational programming of a pro-oxidant/proinflammatory environment that affects the placentas of F2 fetuses. Control and mild pregestational diabetic female rats (F0) were mated with control males, and Idebenone or vehicle was administered to diabetic rats from day 1 of gestation to term. The F1 female offspring were mated with control males and maternal and fetal plasma samples were obtained for metabolic determinations at term. The F2 fetuses and placentas were weighed, and placental protein levels and peroxynitrite-induced damage (immunohistochemistry), mRNA levels (PCR), nitric oxide production (Griess reaction), and number of apoptotic cells (TUNEL) were evaluated. The F1 offspring of F0 diabetic rats (treated or not with Idebenone) developed GDM. The placentas of GDM rats showed a decrease in the mRNA levels of manganese superoxide dismutase and an increase in the production of nitric oxide, peroxynitrite-induced damage, and connective tissue growth factor levels, alterations that were prevented by the maternal Idebenone treatment in F0 rats. In conclusion, the maternal treatment with Idebenone in pregestational diabetic F0 rats ameliorates the pro-oxidant/proinflammatory environment that affects the placentas of F2 fetuses, although it does not prevent F1 rats from developing GDM.


Asunto(s)
Antioxidantes/farmacología , Ubiquinona/análogos & derivados , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Gestacional/metabolismo , Femenino , Macrosomía Fetal/metabolismo , Macrosomía Fetal/fisiopatología , Feto/metabolismo , Masculino , Óxido Nítrico/metabolismo , Placenta/metabolismo , Embarazo , Proteínas Gestacionales , Ratas , Ratas Wistar , Ubiquinona/farmacología
17.
Int J Med Sci ; 18(12): 2653-2660, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34104097

RESUMEN

Background: Macrosomic birth weight has been implicated as a significant risk factor for developing various adult metabolic diseases such as diabetes mellitus and coronary heart diseases; it has also been associated with higher incidences of complicated births. This study aimed to examine the predictability of macrosomic births in hyperglycemic pregnant women using maternal clinical characteristics and serum biomarkers of aneuploidy screening performed in the first half of pregnancy. Methods: A retrospective observational study was performed on a cohort of 1,668 pregnant women who 1) had positive outcomes after undergoing 50-g oral glucose challenge test (OGCT) at two university-based hospitals and 2) underwent any one of the following maternal biomarker screening tests for fetal aneuploidy: triple test, quadruple test, and integrated test. Logistic regression-based models for predicting macrosomic births using maternal characteristics and serum biomarkers were developed and evaluated for prediction power. A nomogram, which is a graphical display of the best predictable model, was then generated. Results: The study cohort included 157 macrosomic birth cases defined as birth weight ≥3,820 g, which was equivalent to the top 10 percentile of the modeling cohort. Three primary models solely based on serum biomarkers achieved area under curves (AUCs) of 0.55-0.62. Expanded models, including maternal demographic and clinical factors, demonstrated an improved performance by 25% (AUCs, 0.69-0.73). Conclusion: Our prediction models will help to identify pregnancies with an elevated risk of macrosomic births in hyperglycemic mothers using maternal clinical factors and serum markers from routine antenatal screening tests. Prediction of macrosomic birth at mid-pregnancy may allow customized antenatal care to reduce the risk of macrosomic births.


Asunto(s)
Peso al Nacer , Diabetes Gestacional/sangre , Macrosomía Fetal/epidemiología , Hiperglucemia/complicaciones , Pruebas de Detección del Suero Materno/estadística & datos numéricos , Adulto , Aneuploidia , Biomarcadores/análisis , Biomarcadores/metabolismo , Glucemia/análisis , Diabetes Gestacional/diagnóstico , Femenino , Macrosomía Fetal/sangre , Macrosomía Fetal/etiología , Macrosomía Fetal/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/sangre , Hiperglucemia/diagnóstico , Hiperglucemia/metabolismo , Recién Nacido , Edad Materna , Embarazo , Estudios Retrospectivos , Factores de Riesgo
18.
Am J Physiol Endocrinol Metab ; 320(6): E1138-E1147, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33938236

RESUMEN

Insulin and insulin-like growth factor-1 (IGF-1) are fetal hormones critical to establishing normal fetal growth. Experimentally elevated IGF-1 concentrations during late gestation increase fetal weight but lower fetal plasma insulin concentrations. We therefore hypothesized that infusion of an IGF-1 analog for 1 wk into late gestation fetal sheep would attenuate fetal glucose-stimulated insulin secretion (GSIS) and insulin secretion in islets isolated from these fetuses. Late gestation fetal sheep received infusions with IGF-1 LR3 (IGF-1, n = 8), an analog of IGF-1 with low affinity for the IGF binding proteins and high affinity for the IGF-1 receptor, or vehicle control (CON, n = 9). Fetal GSIS was measured with a hyperglycemic clamp (IGF-1, n = 8; CON, n = 7). Fetal islets were isolated, and insulin secretion was assayed in static incubations (IGF-1, n = 8; CON, n = 7). Plasma insulin and glucose concentrations in IGF-1 fetuses were lower compared with CON (P = 0.0135 and P = 0.0012, respectively). During the GSIS study, IGF-1 fetuses had lower insulin secretion compared with CON (P = 0.0453). In vitro, glucose-stimulated insulin secretion remained lower in islets isolated from IGF-1 fetuses (P = 0.0447). In summary, IGF-1 LR3 infusion for 1 wk into fetal sheep lowers insulin concentrations and reduces fetal GSIS. Impaired insulin secretion persists in isolated fetal islets indicating an intrinsic islet defect in insulin release when exposed to IGF-1 LR3 infusion for 1 wk. We speculate this alteration in the insulin/IGF-1 axis contributes to the long-term reduction in ß-cell function in neonates born with elevated IGF-1 concentrations following pregnancies complicated by diabetes or other conditions associated with fetal overgrowth.NEW & NOTEWORTHY After a 1-wk infusion of IGF-1 LR3, late gestation fetal sheep had lower plasma insulin and glucose concentrations, reduced fetal glucose-stimulated insulin secretion, and decreased fractional insulin secretion from isolated fetal islets without differences in pancreatic insulin content.


Asunto(s)
Feto/efectos de los fármacos , Glucosa/farmacología , Secreción de Insulina/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Islotes Pancreáticos/efectos de los fármacos , Animales , Diabetes Gestacional/metabolismo , Esquema de Medicación , Femenino , Enfermedades Fetales/metabolismo , Macrosomía Fetal/metabolismo , Macrosomía Fetal/patología , Feto/metabolismo , Edad Gestacional , Bombas de Infusión , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Islotes Pancreáticos/metabolismo , Enfermedades Pancreáticas/metabolismo , Embarazo , Ovinos
19.
Obstet Gynecol Clin North Am ; 48(2): 247-266, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33972064

RESUMEN

Placental regulation of fetal growth involves the integration of multiple signaling pathways that modulate an array of placental functions, including nutrient transport. As a result, the flux of oxygen and nutrients to the fetus is altered, leading to changes in placental and fetal growth. Placental insulin/insulinlike growth factor-1 and mechanistic target of rapamycin signaling and amino acid transport capacity are inhibited in fetal growth restriction and activated in fetal overgrowth, implicating these placental functions in driving fetal growth. With novel approaches to specifically target the placenta, clinical interventions to modulate placental function in high-risk pregnancies can be developed.


Asunto(s)
Desarrollo Fetal , Retardo del Crecimiento Fetal/metabolismo , Macrosomía Fetal/metabolismo , Placenta/metabolismo , Adiponectina/metabolismo , Animales , Diabetes Gestacional/epidemiología , Femenino , Humanos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intercambio Materno-Fetal , Embarazo , Proteínas Gestacionales/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Trofoblastos/metabolismo
20.
Eur J Endocrinol ; 185(1): 77-85, 2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-33914700

RESUMEN

OBJECTIVE: Large birth size programs an elevated risk of type 2 diabetes in adulthood, but data are absent concerning glucose metabolic health impact in infancy. We sought to determine whether the large birth size is associated with insulin resistance and ß-cell function in infancy and evaluate the determinants. DESIGN AND PARTICIPANTS: In the Canadian 3D birth cohort, we conducted a nested matched (1:2) study of 70 large-for-gestational-age (LGA, birth weight >90th percentile) and 140 optimal-for-gestational-age (OGA, 25th-75th percentiles) control infants. The primary outcomes were homeostasis model assessment of insulin resistance (HOMA-IR) and beta-cell function (HOMA-ß) at age 2-years. RESULTS: HOMA-IR and HOMA-ß were similar in LGA and OGA infants. Adjusting for maternal and infant characteristics, decelerated growth in length during early infancy (0-3 months) was associated with a 25.8% decrease (95% confidence intervals 6.7-41.0%) in HOMA-ß. During mid-infancy (3-12 months), accelerated growth in weight was associated with a 25.5% (0.35-56.9%) increase in HOMA-IR, in length with a 69.3% increase (31.4-118.0%) in HOMA-IR and a 24.5% (0.52-54.3%) increase in HOMA-ß. Decelerated growth in length during late infancy (1-2 years) was associated with a 28.4% (9.5-43.4%) decrease in HOMA-IR and a 21.2% (3.9-35.4%) decrease in HOMA-ß. Female sex was associated with higher HOMA-ß, Caucasian ethnicity with lower HOMA-IR, and maternal smoking with lower HOMA-ß. CONCLUSIONS: This study is the first to demonstrate that large birth size is not associated with insulin resistance and ß-cell function in infancy but infancy growth pattern matters. Decelerated infancy growth may be detrimental to beta-cell function.


Asunto(s)
Peso al Nacer , Estatura , Peso Corporal , Desarrollo Infantil , Macrosomía Fetal/metabolismo , Resistencia a la Insulina , Estudios de Casos y Controles , Preescolar , Femenino , Macrosomía Fetal/fisiopatología , Humanos , Lactante , Recién Nacido , Células Secretoras de Insulina/metabolismo , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...