Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Physiol ; 599(5): 1487-1511, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33450094

RESUMEN

KEY POINTS: The prevalence of obesity and non-alcoholic fatty liver disease in children is dramatically increasing at the same time as consumption of foods with a high sugar content. Intake of high fructose corn syrup (HFCS) is a possible aetiology as it is thought to be more lipogenic than glucose. In a mouse model, HFCS intake during adolescence increased fat mass and hepatic lipid levels in male and female mice. However, only males showed impaired glucose tolerance. Multiple metabolites including lipids, bile acids, carbohydrates and amino acids were altered in liver in a sex-specific manner at 6 weeks of age. Some of these changes were also present in adulthood even though HFCS exposure ended at 6 weeks. HFCS significantly altered the gut microbiome, which was associated with changes in key microbial metabolites. These results suggest that HFCS intake during adolescence has profound metabolic changes that are linked to changes in the microbiome and these changes are sex-specific. ABSTRACT: The rapid increase in obesity, diabetes and fatty liver disease in children over the past 20 years has been linked to increased consumption of high fructose corn syrup (HFCS), making it essential to determine the short- and long-term effects of HFCS during this vulnerable developmental window. We hypothesized that HFCS exposure during adolescence significantly impairs hepatic metabolic signalling pathways and alters gut microbial composition, contributing to changes in energy metabolism with sex-specific effects. C57bl/6J mice with free access to HFCS during adolescence (3-6 weeks of age) underwent glucose tolerance and body composition testing and hepatic metabolomics, gene expression and triglyceride content analysis at 6 and 30 weeks of age (n = 6-8 per sex). At 6 weeks HFCS-exposed mice had significant increases in fat mass, glucose intolerance, hepatic triglycerides (females) and de novo lipogenesis gene expression (ACC, DGAT, FAS, ChREBP, SCD, SREBP, CPT and PPARα) with sex-specific effects. At 30 weeks, HFCS-exposed mice also had abnormalities in glucose tolerance (males) and fat mass (females). HFCS exposure enriched carbohydrate, amino acid, long chain fatty acid and secondary bile acid metabolism at 6 weeks with changes in secondary bile metabolism at 6 and 30 weeks. Microbiome studies performed immediately before and after HFCS exposure identified profound shifts of microbial species in male mice only. In summary, short-term HFCS exposure during adolescence induces fatty liver, alters important metabolic pathways, some of which continue to be altered in adulthood, and changes the microbiome in a sex-specific manner.


Asunto(s)
Jarabe de Maíz Alto en Fructosa , Microbiota , Enfermedad del Hígado Graso no Alcohólico , Animales , Femenino , Fructosa , Jarabe de Maíz Alto en Fructosa/efectos adversos , Metabolismo de los Lípidos , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/etiología
2.
Int J Obes (Lond) ; 44(7): 1497-1507, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31852997

RESUMEN

BACKGROUND/OBJECTIVES: Pregnancies complicated by gestational diabetes (GDM) or maternal obesity have been linked to the development of diabetes, obesity, and fatty liver disease later in life with sex-specific manifestations. Alterations in miRNA expression in offspring exposed to GDM and maternal obesity and effects on hepatic development are unknown. Here, we describe how exposure to maternal obesity in utero leads to sex-specific changes in miRNA and target gene expression in human fetal liver. METHODS: Candidate miRNA expression was measured in second trimester amniotic fluid (AF) from women with GDM. Targets of differentially expressed miRNAs were determined and pathway enrichment of target genes was performed. MiRNA and target gene expression were measured in a separate cohort of second trimester primary human fetal hepatocytes (PHFH) exposed to maternal obesity via qPCR and western blot. All studies were IRB approved. RESULTS: GDM-exposed AF had significant increases in miRNAs 199a-3p, 503-5p, and 1268a (fold change (FC) ≥ 1.5, p < 0.05). Female offspring-specific analysis showed enrichment in miRNAs 378a-3p, 885-5p, and 7-1-3p (p < 0.05). MiRNA gene targets were enriched in hepatic pathways. Key genes regulating de novo lipogenesis were upregulated in obesity-exposed PHFH, especially in males. Significantly altered miRNAs in GDM AF were measured in obese-exposed PHFH, with consistent increases in miRNAs 885-5p, 199-3p, 503-5p, 1268a, and 7-1-3p (FC ≥ 1.5, p < 0.05). Female PHFH exposed to maternal obesity had increased expression of miR-885-5p, miR-199-3p, miR-503-5p, miR-1268s, and miR-7-1-3p (p < 0.05), corresponding to decreased target genes expression for ABCA1, PAK4, and INSR. In male PHFHs, no miRNA changes were measured but there was increased expression of ABCA1, PAK4, and INSR (p < 0.05). CONCLUSIONS: Our data suggest sex-specific changes in miRNA and gene expression in PHFH may be one mechanism contributing to the sexual dimorphism of metabolic disease in offspring exposed to GDM and maternal obesity in utero.


Asunto(s)
Diabetes Gestacional , MicroARNs/genética , Obesidad Materna , Caracteres Sexuales , Adulto , Estudios de Casos y Controles , Femenino , Feto , Expresión Génica , Hepatocitos , Humanos , Masculino , Embarazo , Segundo Trimestre del Embarazo
3.
Int J Mol Sci ; 21(3)2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033212

RESUMEN

The placenta is metabolically active and supports the growth of the fetus. We hypothesize that deficits in the capacity of the placenta to maintain bioenergetic and metabolic stability during pregnancy may result in spontaneous preterm birth (SPTB). To explore this hypothesis, we performed a nested cased control study of metabolomic signatures in placentas from women with SPTB (<36 weeks gestation) compared to normal pregnancies (≥38 weeks gestation). To control for the effects of gestational age on placenta metabolism, we also studied a subset of metabolites in non-laboring preterm and term Rhesus monkeys. Comprehensive quantification of metabolites demonstrated a significant elevation in the levels of amino acids, prostaglandins, sphingolipids, lysolipids, and acylcarnitines in SPTB placenta compared to term placenta. Additional quantification of placental acylcarnitines by tandem mass spectrometry confirmed the significant elevation in SPTB human, with no significant differences between midgestation and term placenta in Rhesus macaque. Fatty acid oxidation as measured by the flux of 3H-palmitate in SPTB placenta was lower than term. Collectively, significant and biologically relevant alterations in the placenta metabolome were identified in SPTB placenta. Altered acylcarnitine levels and fatty acid oxidation suggest that disruption in normal substrate metabolism is associated with SPTB.


Asunto(s)
Placenta/metabolismo , Nacimiento Prematuro/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Feto/metabolismo , Edad Gestacional , Humanos , Recién Nacido , Metabolómica/métodos , Embarazo
4.
Environ Res ; 176: 108558, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31271921

RESUMEN

BACKGROUND: Perfluorooctanoate (PFOA) has been used extensively in the manufacture of both commercial and household products. PFOA serum concentrations have been associated with adverse health effects, including lower body mass in children and infants. OBJECTIVE: To determine if there is an association between serum PFOA concentration and body mass, serum insulin and lipid profile in exposed young girls. METHODS: We conducted a cross-sectional study of PFAS environmental biomarkers and insulin resistance in 6 to 8 year-old girls from Greater Cincinnati (n=353). In 2004-2006, blood samples were obtained to measure polyfluoroalkyl substances (PFAS), fasting insulin, glucose and lipids. Clinical exams included anthropometric measurements and pubertal maturation staging. Linear regression and mediation analyses, specifically structural equation modeling (SEM), were used to determine the strength and direction of the relationships between PFAS, pubertal maturation status, body mass index (BMI), cholesterol and insulin resistance. RESULTS: The median PFOA (7.7ng/ml) was twice the National Health and Nutrition Examination Survey (2005-2006). Only PFOA, a PFAS sub-species, showed statistically significant relationships with the outcomes. In regression models, PFOA was associated with decreased BMI and waist-to-height ratio (p=0.0008; p=0.0343), HDL-cholesterol (p=0.0046) and had a borderline inverse association with the HOMA Index of insulin resistance (p=0.0864). In SEM, PFOA retained an inverse relationship with BMI (p<0.0001) but the relationships with HOMA and HDL-cholesterol were no longer statistically significant. Pubertal initiation (Tanner breast or pubic stage 2 or greater) and BMI were associated with increased HOMA Index (p<0.0001). CONCLUSIONS: These findings suggest PFOA exposure in young girls affects both BMI and ultimately insulin resistance. In mediation analysis with puberty in the model, the direct effects of PFOA on insulin resistance and were reduced.


Asunto(s)
Índice de Masa Corporal , Caprilatos , Exposición a Riesgos Ambientales/estadística & datos numéricos , Contaminantes Ambientales/análisis , Fluorocarburos , Resistencia a la Insulina , Lípidos/sangre , Niño , Estudios Transversales , Femenino , Humanos , Insulina , Encuestas Nutricionales , Maduración Sexual
5.
Int J Mol Sci ; 19(9)2018 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201937

RESUMEN

Maternal diabetes and obesity induce marked abnormalities in glucose homeostasis and insulin secretion in the fetus, and are linked to obesity, diabetes, and metabolic disease in the offspring, with specific metabolic characterization based on offspring sex. Gestational diabetes (GDM) has profound effects on the intrauterine milieu, which may reflect and/or modulate the function of the maternal⁻fetal unit. In order to characterize metabolic factors that affect offspring development, we profiled the metabolome of second trimester amniotic fluid (AF) from women who were subsequently diagnosed with gestational diabetes (GDM) using a targeted metabolomics approach, profiling 459 known biochemicals through gas chromatography/mass spectrometry (GC/MS) and liquid chromatography/mass spectrometry (LC/MS) assays. Using a nested case-control study design, we identified 69 total biochemicals altered by GDM exposure, while sex-specific analysis identified 44 and 58 metabolites in male and female offspring, respectively. The most significant changes were in glucose, amino acid, glutathione, fatty acid, sphingolipid, and bile acid metabolism with specific changes identified based on the offspring sex. Targeted isotope dilution LC/MS confirmatory assays measured significant changes in docosahexaenoic acid and arachidonic acid. We conclude that the sex-specific alterations in GDM maternal⁻fetal metabolism may begin to explain the sex-specific metabolic outcomes seen in offspring exposed to GDM in utero.


Asunto(s)
Líquido Amniótico/metabolismo , Diabetes Gestacional/metabolismo , Metabolómica/métodos , Segundo Trimestre del Embarazo/metabolismo , Adulto , Ácido Araquidónico/análisis , Estudios de Casos y Controles , Cromatografía Liquida , Ácidos Docosahexaenoicos/análisis , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Recién Nacido , Masculino , Espectrometría de Masas , Embarazo , Factores Sexuales
6.
Pediatr Diabetes ; 18(3): 167-177, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28401680

RESUMEN

Although the factors responsible for the recent increase in the prevalence of diabetes worldwide are not entirely known, the morbidity associated with this disease results in substantial health and economic burden on society. Epigenetic modifications, including DNA methylation have been identified as one mechanism by which the environment interacts with the genome and there is evidence that alterations in DNA methylation may contribute to the increased prevalence of both type 1 and type 2 diabetes. This review provides a summary of DNA methylation and its role in gene regulation, and includes descriptions of various techniques to measure site-specific and genome-wide DNA methylation changes. In addition, we review current literature highlighting the complex relationship between DNA methylation, gene expression, and the development of diabetes and related complications. In studies where both DNA methylation and gene expression changes were reported, DNA methylation status had a strong inverse correlation with gene expression, suggesting that this interaction may be a potential future therapeutic target. We highlight the emerging use of genome-wide DNA methylation profiles as a biomarker to predict patients at risk of developing diabetes or specific complications of diabetes. The development of a predictive model that incorporates both genetic sequencing and DNA methylation data may be an effective diagnostic approach for all types of diabetes and could lead to additional innovative therapies.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Animales , Biomarcadores/metabolismo , Terapia Combinada , Complicaciones de la Diabetes/epidemiología , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/terapia , Transición de la Salud , Humanos , Resistencia a la Insulina , Factores de Riesgo
7.
Drug Discov Today Dis Models ; 10(2): e71-e77, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24949076

RESUMEN

Intrauterine growth retardation has been linked to the development of type 2 diabetes later in life and the mechanisms underlying this phenomena are unknown. Epidemiological studies in humans show a distinct link with the exposure to an intrauterine insult that results in low birth weight and the development of type 2 diabetes in adulthood. Intrauterine growth retardation can be induced in rodent models by exposing the pregnant rat to a low protein diet, total calorie restriction, high dose glucocorticoids or inducing uteroplacental insufficiency, all which result in abnormalities in glucose homeostasis in the offspring later in life. Animal models of intrauterine growth retardation allow for a better characterization of changes in glucose homeostasis and corresponding changes in gene expression that can provide insight in the mechanisms by which intrauterine growth retardation leads to type 2 diabetes.

8.
Curr Diab Rep ; 12(1): 67-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22127642

RESUMEN

The link between an adverse intrauterine environment and the development of disease later in life has been observed in offspring of pregnancies complicated by obesity and diabetes, but the molecular mechanisms underlying this phenomenon are unknown. In this review, we highlight recent publications exploring the role of gestational diabetes mellitus in the programming of disease in the offspring. We also review recent publications aiming to identify mechanisms responsible for the "programming effect" that results from exposure to diabetes in utero. Finally, we highlight research on the role of epigenetic regulation of gene expression in an animal model of uteroplacental insufficiency where the offspring develop diabetes as a model by which an exposure to the mother can alter epigenetic modifications that affect expression of key genes and ultimately lead to the development of diabetes in the offspring.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/genética , Epigénesis Genética , Retardo del Crecimiento Fetal/genética , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal , Adolescente , Animales , Niño , Preescolar , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Gestacional/epidemiología , Femenino , Retardo del Crecimiento Fetal/epidemiología , Humanos , Lactante , Recién Nacido , Masculino , Obesidad/epidemiología , Embarazo , Ratas , Estados Unidos/epidemiología
9.
BMJ Case Rep ; 15(1)2022 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-34980640

RESUMEN

Pseudohypoaldosteronism type 1B is a rare autosomal recessive disorder caused by dysfunction of amiloride-sensitive epithelial sodium channels (ENaCs). We present the case of a neonate with cardiogenic shock after cardiac arrest due to profound hyperkalaemia. Genetic testing revealed a novel homozygous variant in SCNNIA We review diagnostic considerations including the molecular mechanisms of disease, discuss treatment approaches and highlight the possible significance of the diversity of pulmonary ENaCs.


Asunto(s)
Hiperpotasemia , Seudohipoaldosteronismo , Amilorida , Canales Epiteliales de Sodio/genética , Homocigoto , Humanos , Hiperpotasemia/diagnóstico , Hiperpotasemia/etiología , Recién Nacido , Seudohipoaldosteronismo/complicaciones , Seudohipoaldosteronismo/diagnóstico , Seudohipoaldosteronismo/genética
10.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894232

RESUMEN

Intrauterine growth restriction (IUGR) leads to the development of type 2 diabetes in adulthood, and the permanent alterations in gene expression implicate an epigenetic mechanism. Using a rat model of IUGR, we performed TrueSeq-HELP Tagging to assess the association of DNA methylation changes and gene dysregulation in islets. We identified 511 differentially methylated regions (DMRs) and 4377 significantly altered single CpG sites. Integrating the methylome and our published transcriptome data sets resulted in the identification of pathways critical for islet function. The identified DMRs were enriched with transcription factor binding motifs, such as Elk1, Etv1, Foxa1, Foxa2, Pax7, Stat3, Hnf1, and AR. In silico analysis of 3-dimensional chromosomal interactions using human pancreas and islet Hi-C data sets identified interactions between 14 highly conserved DMRs and 35 genes with significant expression changes at an early age, many of which persisted in adult islets. In adult islets, there were far more interactions between DMRs and genes with significant expression changes identified with Hi-C, and most of them were critical to islet metabolism and insulin secretion. The methylome was integrated with our published genome-wide histone modification data sets from IUGR islets, resulting in further characterization of important regulatory regions of the genome altered by IUGR containing both significant changes in DNA methylation and specific histone marks. We identified novel regulatory regions in islets after exposure to IUGR, suggesting that epigenetic changes at key transcription factor binding motifs and other gene regulatory regions may contribute to gene dysregulation and an abnormal islet phenotype in IUGR rats.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética , Retardo del Crecimiento Fetal/genética , Regulación de la Expresión Génica , Islotes Pancreáticos/metabolismo , Animales , Sitios de Unión , Islas de CpG , Diabetes Mellitus Tipo 2/genética , Femenino , Estudio de Asociación del Genoma Completo , Histonas/química , Humanos , Islotes Pancreáticos/química , Islotes Pancreáticos/embriología , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/metabolismo
11.
J Clin Invest ; 118(8): 2877-86, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18596924

RESUMEN

Congenital hyperinsulinism is a condition of dysregulated insulin secretion often caused by inactivating mutations of the ATP-sensitive K+ (KATP) channel in the pancreatic beta cell. Though most disease-causing mutations of the 2 genes encoding KATP subunits, ABCC8 (SUR1) and KCNJ11 (Kir6.2), are recessively inherited, some cases of dominantly inherited inactivating mutations have been reported. To better understand the differences between dominantly and recessively inherited inactivating KATP mutations, we have identified and characterized 16 families with 14 different dominantly inherited KATP mutations, including a total of 33 affected individuals. The 16 probands presented with hypoglycemia at ages from birth to 3.3 years, and 15 of 16 were well controlled on diazoxide, a KATP channel agonist. Of 29 adults with mutations, 14 were asymptomatic. In contrast to a previous report of increased diabetes risk in dominant KATP hyperinsulinism, only 4 of 29 adults had diabetes. Unlike recessive mutations, dominantly inherited KATP mutant subunits trafficked normally to the plasma membrane when expressed in COSm6 cells. Dominant mutations also resulted in different channel-gating defects, as dominant ABCC8 mutations diminished channel responses to magnesium adenosine diphosphate or diazoxide, while dominant KCNJ11 mutations impaired channel opening, even in the absence of nucleotides. These data highlight distinctive features of dominant KATP hyperinsulinism relative to the more common and more severe recessive form, including retention of normal subunit trafficking, impaired channel activity, and a milder hypoglycemia phenotype that may escape detection in infancy and is often responsive to diazoxide medical therapy, without the need for surgical pancreatectomy.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Hiperinsulinismo Congénito/genética , Hipoglucemia/genética , Canales KATP/genética , Mutación , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/genética , Adenosina Difosfato/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células COS , Chlorocebus aethiops , Diazóxido/uso terapéutico , Femenino , Genes Dominantes , Prueba de Tolerancia a la Glucosa , Heterocigoto , Humanos , Hipoglucemia/complicaciones , Hipoglucemia/terapia , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Masculino , Persona de Mediana Edad , Técnicas de Placa-Clamp , Linaje , Receptores de Sulfonilureas
12.
Endocrinol Metab Clin North Am ; 49(4): 695-723, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33153675

RESUMEN

Although type 1 diabetes mellitus and, to a lesser extent, type 2 diabetes mellitus, are the prevailing forms of diabetes in youth, atypical forms of diabetes are not uncommon and may require etiology-specific therapies. By some estimates, up to 6.5% of children with diabetes have monogenic forms. Mitochondrial diabetes and cystic fibrosis related diabetes are less common but often noted in the underlying disease. Atypical diabetes should be considered in patients with a known disorder associated with diabetes, aged less than 25 years with nonautoimmune diabetes and without typical characteristics of type 2 diabetes mellitus, and/or with comorbidities associated with atypical diabetes.


Asunto(s)
Fibrosis Quística , Diabetes Mellitus , Enfermedades Mitocondriales , Adolescente , Adulto , Niño , Preescolar , Fibrosis Quística/complicaciones , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/etiología , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Humanos , Lactante , Recién Nacido , Enfermedades Mitocondriales/complicaciones , Adulto Joven
13.
J Clin Endocrinol Metab ; 105(2)2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31536135

RESUMEN

CONTEXT: Prenatal exposure to bisphenol A (BPA) is linked to obesity and diabetes but the molecular mechanisms driving these phenomena are not known. Alterations in deoxyribonucleic acid (DNA) methylation in amniocytes exposed to BPA in utero represent a potential mechanism leading to metabolic dysfunction later in life. OBJECTIVE: To profile changes in genome-wide DNA methylation and expression in second trimester human amniocytes exposed to BPA in utero. DESIGN: A nested case-control study was performed in amniocytes matched for offspring sex, maternal race/ethnicity, maternal age, gestational age at amniocentesis, and gestational age at birth. Cases had amniotic fluid BPA measuring 0.251 to 23.74 ng/mL. Sex-specific genome-wide DNA methylation analysis and RNA-sequencing (RNA-seq) were performed to determine differentially methylated regions (DMRs) and gene expression changes associated with BPA exposure. Ingenuity pathway analysis was performed to identify biologically relevant pathways enriched after BPA exposure. In silico Hi-C analysis identified potential chromatin interactions with DMRs. RESULTS: There were 101 genes with altered expression in male amniocytes exposed to BPA (q < 0.05) in utero, with enrichment of pathways critical to hepatic dysfunction, collagen signaling and adipogenesis. Thirty-six DMRs were identified in male BPA-exposed amniocytes and 14 in female amniocyte analysis (q < 0.05). Hi-C analysis identified interactions between DMRs and 24 genes with expression changes in male amniocytes and 12 in female amniocytes (P < 0.05). CONCLUSION: In a unique repository of human amniocytes exposed to BPA in utero, sex-specific analyses identified gene expression changes in pathways associated with metabolic disease and novel DMRs with potential distal regulatory functions.


Asunto(s)
Amnios/citología , Compuestos de Bencidrilo/efectos adversos , Epigenoma/efectos de los fármacos , Exposición Materna/efectos adversos , Fenoles/efectos adversos , Factores Sexuales , Transcriptoma/efectos de los fármacos , Amnios/efectos de los fármacos , Amnios/embriología , Estudios de Casos y Controles , Metilación de ADN/efectos de los fármacos , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Obesidad/inducido químicamente , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Análisis de Secuencia de ARN
14.
J Clin Endocrinol Metab ; 105(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32687192

RESUMEN

CONTEXT: Gestational diabetes (GDM) has profound effects on the intrauterine metabolic milieu and is linked to obesity and diabetes in offspring, but the mechanisms driving these effects remain largely unknown. Alterations in DNA methylation and gene expression in amniocytes exposed to GDM in utero represent a potential mechanism leading to metabolic dysfunction later in life. OBJECTIVE: To profile changes in genome-wide DNA methylation and expression in human amniocytes exposed to GDM. DESIGN: A nested case-control study (n = 14 pairs) was performed in amniocytes matched for offspring sex, maternal race/ethnicity, maternal age, gestational age at amniocentesis, and gestational age at birth. Sex-specific genome-wide DNA methylation analysis and RNA-sequencing were completed and differentially methylated regions (DMRs) and gene expression changes were identified. Ingenuity pathway analysis identified biologically relevant pathways enriched after GDM exposure. In silico high-throughput chromosome conformation capture (Hi-C) analysis identified potential chromatin interactions with DMRs. RESULTS: Expression of interferon-stimulated genes was increased in GDM amniocytes, accounting for 6 of the top 10 altered genes (q < 0.05). Enriched biological pathways in GDM amniocytes included pathways involving inflammation, the interferon response, fatty liver disease, monogenic diabetes, and atherosclerosis. Forty-two DMRs were identified in male GDM-exposed amniocytes and 20 in female amniocyte analysis (q < 0.05). Hi-C analysis identified interactions between DMRs and 11 genes with significant expression changes in male amniocytes and 9 in female amniocytes (P < .05). CONCLUSION: In a unique repository of human amniocytes exposed to GDM in utero, transcriptome analysis identified enrichment of inflammation and interferon-related pathways and novel DMRs with potential distal regulatory functions.


Asunto(s)
Líquido Amniótico/metabolismo , Diabetes Gestacional/metabolismo , Epigénesis Genética/inmunología , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal/genética , Adulto , Líquido Amniótico/citología , Líquido Amniótico/inmunología , Peso al Nacer/genética , Estudios de Casos y Controles , Cromatina/metabolismo , Islas de CpG/genética , Metilación de ADN , Epigenoma , Femenino , Edad Gestacional , Humanos , Recién Nacido , Interferones/inmunología , Interferones/metabolismo , Masculino , Edad Materna , Obesidad/inmunología , Obesidad/metabolismo , Embarazo , Segundo Trimestre del Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , RNA-Seq , Factores Sexuales , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma
15.
PLoS One ; 13(2): e0190698, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29470513

RESUMEN

AIMS/HYPOTHESIS: We hypothesized that diabetes during pregnancy (DDP) alters genome-wide DNA methylation in placenta resulting in differentially methylated loci of metabolically relevant genes and downstream changes in RNA and protein expression. METHODS: We mapped genome-wide DNA methylation with the Infinium 450K Human Methylation Bead Chip in term fetal placentae from Native American and Hispanic women with DDP using a nested case-control design (n = 17 pairs). RNA expression and protein levels were assayed via RNA-Seq and Western Blot. RESULTS: Genome-wide DNA methylation analysis revealed 465 CpG sites with significant changes for male offspring, 247 for female offspring, and 277 for offspring of both sexes (p<0.001). Placentae from female offspring were 40% more likely to have significant gains in DNA methylation compared with placentae from male offspring exposed to DDP (p<0.001). Changes in DNA methylation corresponded to changes in RNA and protein levels for 6 genes: PIWIL3, CYBA, GSTM1, GSTM5, KCNE1 and NXN. Differential DNA methylation was detected at loci related to mitochondrial function, DNA repair, inflammation, oxidative stress. CONCLUSIONS/INTERPRETATION: These findings begin to explain mechanisms responsible for the increased risk for obesity and type 2 diabetes in offspring of mothers with DDP.


Asunto(s)
Metilación de ADN , Expresión Génica , Placenta/metabolismo , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/metabolismo , Adulto , Estudios de Casos y Controles , Islas de CpG , Diabetes Mellitus Tipo 2/etiología , Femenino , Humanos , Recién Nacido , Masculino , Obesidad/etiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores Sexuales , Adulto Joven
16.
Reprod Toxicol ; 67: 1-9, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27829162

RESUMEN

Bisphenol A (BPA) is an endocrine disrupting chemical with ubiquitous environmental exposure. Animal studies have demonstrated that in utero BPA exposure leads to increased adult body weight. Our aim was to characterize human fetal BPA exposure by measuring BPA concentration in second trimester amniotic fluid (AF) samples and to study its relationship with birth weight (BW) in full term infants. To achieve these goals, we developed a total BPA assay utilizing derivatization with pentafluorobenzyl followed by analysis with LC-ECAPCI-MS/MS with a limit of detection of 0.08ng/mL and limit of quantification (LOQ) of 0.25ng/mL. The mean BW of infants with AF BPA 0.40-2.0ng/mL was 241.8g less than infants with AF BPA less than the LOQ after controlling for covariates (p=0.049). No effect was seen outside this range indicating a non-monotonic effect. Our data suggest that low level BPA exposure in utero decreases BW and needs further study.


Asunto(s)
Líquido Amniótico/química , Compuestos de Bencidrilo/análisis , Disruptores Endocrinos/análisis , Recién Nacido de Bajo Peso , Fenoles/análisis , Efectos Tardíos de la Exposición Prenatal/etiología , Cromatografía Liquida , Femenino , Humanos , Límite de Detección , Embarazo , Segundo Trimestre del Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Espectrometría de Masas en Tándem
17.
Biology (Basel) ; 3(4): 739-51, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25393317

RESUMEN

Although CpG dinucleotides remain the primary site for DNA methylation in mammals, there is emerging evidence that DNA methylation at non-CpG sites (CpA, CpT and CpC) is not only present in mammalian cells, but may play a unique role in the regulation of gene expression. For some time it has been known that non-CpG methylation is abundant in plants and present in mammalian embryonic stem cells, but non-CpG methylation was thought to be lost upon cell differentiation. However, recent publications have described a role for non-CpG methylation in adult mammalian somatic cells including the adult mammalian brain, skeletal muscle, and hematopoietic cells and new interest in this field has been stimulated by the availability of high throughput sequencing techniques that can accurately measure this epigenetic modification. Genome wide assays indicate that non-CpG methylation is negligible in human fetal brain, but abundant in human adult brain tissue. Genome wide measurement of non-CpG methylation coupled with RNA-Sequencing indicates that in the human adult brain non-CpG methylation levels are inversely proportional to the abundance of mRNA transcript at the associated gene. Additionally specific examples where alterations in non-CpG methylation lead to changes in gene expression have been described; in PGC1α in human skeletal muscle, IFN-γ in human T-cells and SYT11 in human brain, all of which contribute to the development of human disease.

18.
Horm Res Paediatr ; 80(1): 18-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23859901

RESUMEN

BACKGROUND/AIMS: In a family with congenital hyperinsulinism (HI), first described in the 1950s by McQuarrie, we examined the genetic locus and clinical phenotype of a novel form of dominant HI. METHODS: We surveyed 25 affected individuals, 7 of whom participated in tests of insulin dysregulation (24-hour fasting, oral glucose and protein tolerance tests). To identify the disease locus and potential disease-associated mutations we performed linkage analysis, whole transcriptome sequencing, whole genome sequencing, gene capture, and next generation sequencing. RESULTS: Most affecteds were diagnosed with HI before age one and 40% presented with a seizure. All affecteds responded well to diazoxide. Affecteds failed to adequately suppress insulin secretion following oral glucose tolerance test or prolonged fasting; none had protein-sensitive hypoglycemia. Linkage analysis mapped the HI locus to Chr10q21-22, a region containing 48 genes. Three novel noncoding variants were found in hexokinase 1 (HK1) and one missense variant in the coding region of DNA2. CONCLUSION: Dominant, diazoxide-responsive HI in this family maps to a novel locus on Chr10q21-22. HK1 is the more attractive disease gene candidate since a mutation interfering with the normal suppression of HK1 expression in beta-cells could readily explain the hypoglycemia phenotype of this pedigree.


Asunto(s)
Cromosomas Humanos Par 10/genética , Hiperinsulinismo Congénito/genética , Genes Dominantes , Hexoquinasa/genética , Adulto , Anciano de 80 o más Años , Glucemia/metabolismo , Preescolar , Hiperinsulinismo Congénito/tratamiento farmacológico , Diazóxido/uso terapéutico , Ayuno , Femenino , Ligamiento Genético , Humanos , Lactante , Insulina/metabolismo , Secreción de Insulina , Masculino , Persona de Mediana Edad , Mutación , Análisis de Secuencia de ADN
19.
J Clin Endocrinol Metab ; 96(7): 1960-5, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21490072

RESUMEN

OBJECTIVE: The aim was to describe the clinical presentation and to characterize the genetic mutation present in a child with congenital malabsorptive diarrhea and neonatal diabetes. RESEARCH DESIGN AND METHODS: Clinical data were obtained from chart review. Histopathological characterization of intestinal samples and neurogenin-3 (NEUROG3) sequencing were performed. Expression and function of the mutated NEUROG3 protein were assessed by Western blot analysis and luciferase reporter assay. RESULTS: At birth, the proband was small for gestational age. She presented for evaluation with persistent diarrhea and a poor postnatal growth pattern. Although the pancreas was present, serum amylase and fecal elastase levels were decreased, and blood glucose levels were persistently elevated by 5 months of age. Immunostaining of a small intestine biopsy for chromogranin A demonstrated complete absence of neuroendocrine cells. Genetic analysis revealed a nonsense mutation (E123X) in the region encoding helix II of the NEUROG3 gene, leading to premature termination at amino acid 123. The mutated truncated NEUROG3 protein was identified by Western blot analysis. Reporter assays show decreased transactivation of the NEUROD1 promoter by mutant NEUROG3 protein as compared to wild type. CONCLUSIONS: This report describes a newly identified nonsense mutation in human NEUROG3 that in the homozygous state is associated with neonatal diabetes and malabsorptive diarrhea.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diabetes Mellitus/genética , Diarrea/genética , Proteínas del Tejido Nervioso/genética , Diabetes Mellitus/congénito , Diabetes Mellitus/patología , Diarrea/congénito , Diarrea/patología , Femenino , Humanos , Lactante , Recién Nacido , Intestino Delgado/patología , Mutación
20.
Trends Endocrinol Metab ; 21(4): 223-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19864158

RESUMEN

Type 2 diabetes (T2D) is a disorder of complex genetics influenced by interactions between susceptible genetic loci and environmental perturbations. Intrauterine growth retardation is one such environmental perturbation linked to the development of T2D in adulthood. An abnormal metabolic intrauterine milieu affects fetal development by permanently modifying expression of key genes regulating beta-cell development (Pdx1) and glucose transport (Glut4) in muscle. Epigenetic regulation of gene expression is one mechanism by which genetic susceptibility and environmental insults can lead to T2D. Therefore, therapeutic agents targeting epigenetic gene regulation can ultimately be used to treat T2D; however, there is much to be learned about genome-wide epigenetic programming of health and disease before these therapies can be used in patient care.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Epigénesis Genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Diabetes Mellitus Tipo 2/etiología , Retardo del Crecimiento Fetal/genética , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Estrés Oxidativo , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA