Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Clin Sci (Lond) ; 138(18): 1179-1207, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39289953

RESUMEN

With the global prevalence of diabetes increasing, more people of reproductive age are experiencing hyperglycaemic pregnancies. Maternal Type 1 (T1DM) or Type 2 (T2DM) diabetes mellitus, and gestational diabetes mellitus (GDM) are associated with maternal cardiovascular and metabolic complications. Pregnancies complicated by maternal diabetes also increase the risk of short- and long-term health complications for the offspring, including altered fetal growth and the onset of T2DM and cardiometabolic diseases throughout life. Despite advanced methods for improving maternal glucose control, the prevalence of adverse maternal and offspring outcomes associated with maternal diabetes remains high. The placenta is a key organ at the maternal-fetal interface that regulates fetal growth and development. In pregnancies complicated by maternal diabetes, altered placental development and function has been linked to adverse outcomes in both mother and fetus. Emerging evidence suggests that microRNAs (miRNAs) are key molecules involved in mediating these changes. In this review, we describe the role of miRNAs in normal pregnancy and discuss how miRNA dysregulation in the placenta and maternal circulation is associated with suboptimal placental development and pregnancy outcomes in individuals with maternal diabetes. We also discuss evidence demonstrating that miRNA dysregulation may affect the long-term health of mothers and their offspring. As such, miRNAs are potential candidates as biomarkers and therapeutic targets in diabetic pregnancies at risk of adverse outcomes.


Asunto(s)
Diabetes Gestacional , MicroARNs , Placenta , Embarazo en Diabéticas , Humanos , Embarazo , Femenino , MicroARNs/genética , MicroARNs/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/metabolismo , Placenta/metabolismo , Resultado del Embarazo
2.
Am J Obstet Gynecol ; 230(2): 254.e1-254.e13, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37531989

RESUMEN

BACKGROUND: Hyperglycemia from pregestational diabetes mellitus induces neural tube defects in the developing fetus. Folate supplementation is the only effective way to prevent neural tube defects; however, some cases of neural tube defects are resistant to folate. Excess folate has been linked to higher maternal cancer risk and infant allergy. Therefore, additional interventions are needed. Understanding the mechanisms underlying maternal diabetes mellitus-induced neural tube defects can identify potential targets for preventing such defects. Despite not yet being in clinical use, growing evidence suggests that microRNAs are important intermediates in embryonic development and can serve as both biomarkers and drug targets for disease intervention. Our previous studies showed that maternal diabetes mellitus in vivo activates the inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in the developing embryo and that a high glucose condition in vitro reduces microRNA-322 (miR-322) levels. IRE1α is an RNA endonuclease; however, it is unknown whether IRE1α targets and degrades miR-322 specifically or whether miR-322 degradation leads to neural tube defects via apoptosis. We hypothesize that IRE1α can inhibit miR-322 in maternal diabetes mellitus-induced neural tube defects and that restoring miR-322 expression in developing neuroepithelium ameliorates neural tube defects. OBJECTIVE: This study aimed to identify potential targets for preventing maternal diabetes mellitus-induced neural tube defects and to investigate the roles and relationship of a microRNA and an RNA endonuclease in mouse embryos exposed to maternal diabetes mellitus. STUDY DESIGN: To determine whether miR-322 reduction is necessary for neural tube defect formation in pregnancies complicated by diabetes mellitus, male mice carrying a transgene expressing miR-322 were mated with nondiabetic or diabetic wide-type female mice to generate embryos with or without miR-322 overexpression. At embryonic day 8.5 when the neural tube is not yet closed, embryos were harvested for the assessment of 3 miR-322 transcripts (primary, precursor, and mature miR-322), tumor necrosis factor receptor-associated factor 3 (TRAF3), and neuroepithelium cell survival. Neural tube defect incidences were determined in embryonic day 10.5 embryos when the neural tube should be closed if there is no neural tube defect formation. To identify which miR-322 transcript is affected by maternal diabetes mellitus and high glucose conditions, 3 miR-322 transcripts were assessed in embryos from dams with or without diabetes mellitus and in C17.2 mouse neural stem cells treated with different concentrations of glucose and at different time points. To determine whether the endonuclease IRE1α targets miR-322, small interfering RNA knockdown of IRE1α or overexpression of inositol-requiring transmembrane kinase/endoribonuclease 1α by DNA plasmid transfection was used to determine the effect of IRE1α deficiency or overexpression on miR-322 expression. RNA immunoprecipitation was performed to reveal the direct targets of inositol-requiring transmembrane kinase/endoribonuclease 1α. RESULTS: Maternal diabetes mellitus suppressed miR-322 expression in the developing neuroepithelium. Restoring miR-322 expression in the neuroepithelium blocked maternal diabetes mellitus-induced caspase-3 and caspase-8 cleavage and cell apoptosis, leading to a neural tube defect reduction. Reversal of maternal diabetes mellitus-inhibited miR-322 via transgenic overexpression prevented TRAF3 up-regulation in embryos exposed to maternal diabetes mellitus. Activated IRE1α acted as an endonuclease and degraded precursor miR-322, resulting in mature miR-322 reduction. CONCLUSION: This study supports the crucial role of the IRE1α-microRNA-TRAF3 circuit in the induction of neuroepithelial cell apoptosis and neural tube defect formation in pregnancies complicated by diabetes mellitus and identifies IRE1α and miR-322 as potential targets for preventing maternal diabetes mellitus-induced neural tube defects.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Gestacional , MicroARNs , Defectos del Tubo Neural , Embarazo en Diabéticas , Humanos , Embarazo , Masculino , Femenino , Ratones , Animales , MicroARNs/genética , MicroARNs/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/metabolismo , Diabetes Gestacional/genética , Glucosa , Ácido Fólico , Inositol
3.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38003449

RESUMEN

Congenital heart defects (CHDs) are the most common form of birth defects in humans. They occur in 9 out of 1000 live births and are defined as structural abnormalities of the heart. Understanding CHDs is difficult due to the heterogeneity of the disease and its multifactorial etiology. Advances in genomic sequencing have made it possible to identify the genetic factors involved in CHDs. However, genetic origins have only been found in a minority of CHD cases, suggesting the contribution of non-inherited (environmental) risk factors to the etiology of CHDs. Maternal pregestational diabetes is associated with a three- to five-fold increased risk of congenital cardiopathies, but the underlying molecular mechanisms are incompletely understood. According to current hypotheses, hyperglycemia is the main teratogenic agent in diabetic pregnancies. It is thought to induce cell damage, directly through genetic and epigenetic dysregulations and/or indirectly through production of reactive oxygen species (ROS). The purpose of this review is to summarize key findings on the molecular mechanisms altered in cardiac development during exposure to hyperglycemic conditions in utero. It also presents the various in vivo and in vitro techniques used to experimentally model pregestational diabetes. Finally, new approaches are suggested to broaden our understanding of the subject and develop new prevention strategies.


Asunto(s)
Diabetes Gestacional , Cardiopatías Congénitas , Hiperglucemia , Embarazo en Diabéticas , Embarazo , Femenino , Humanos , Diabetes Gestacional/genética , Factores de Riesgo , Cardiopatías Congénitas/genética , Embarazo en Diabéticas/genética , Hiperglucemia/complicaciones , Hiperglucemia/genética
4.
Mol Cell Biochem ; 477(5): 1629-1643, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35229243

RESUMEN

Precise differentiation of glucokinase (GCK) monogenic diabetes from gestational diabetes mellitus (GDM) is critical for accurate management of the pregnancy outcome. We screened GCK-MODY complicating pregnancies in Chinese GDM patients, explored the pathogenesis of novel GCK mutations, and evaluated the patients' pregnancy outcome and management. The GCK gene from 411 GDM patients was screened with PCR-direct sequencing and multiplex ligation-dependent probe amplification (MLPA) and 15 GCK mutations were identified. We also retrospectively analyzed a total of 65 pregnancies from 21 GCK-MODY families, wherein 41 were from 15 maternal families and 24 were from six paternal families. Bioinformatic analysis and biochemical functional study were conducted to identify novel GCK mutations. In total, we identified 21 GCK mutations: 15 from the 411 GDM patients and six from 24 fathers. Of th Asp78Asn (GAC → AAC), Met87Arg (ATG → AGG), Leu451Val (CTT → GTT), Leu451Pro (CTG → CCG) and 1019 + 20G > A e mutations, five, i.e., were novel and deleterious, with markedly decreased enzyme activity and thermal stability. The unaffected offspring of GCK mutation-affected mothers were heavier than affected offspring (p < 0.001). Of 21 insulin-treated affected mothers, 10 had maternal hypoglycemia (47.6%) and seven had perinatal complications (33.3%), and the affected offspring of the insulin-treated affected mothers had significantly lower birth weights than that of the 20 diet-control affected mothers (p = 0.031). In this study, the prevalence of GCK-MODY complicating pregnancy in Chinese GDM patients was 3.6% (15/411). The defective GCK may contribute to the hyperglycemia in GCK-MODY. Insulin therapy is not beneficial for GCK-MODY complicating pregnancy and therefore should not be recommended.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Embarazo en Diabéticas , China , Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/genética , Femenino , Glucoquinasa/genética , Humanos , Insulina/genética , Mutación , Embarazo , Resultado del Embarazo , Embarazo en Diabéticas/epidemiología , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/terapia , Estudios Retrospectivos
5.
Genesis ; 59(11): e23449, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34498806

RESUMEN

Congenital heart disease (CHD) is the leading cause of birth defect-related death in infants and is a global pediatric health concern. While the genetic causes of CHD have become increasingly recognized with advances in genome sequencing technologies, the etiology for the majority of cases of CHD is unknown. The maternal environment during embryogenesis has a profound impact on cardiac development, and numerous environmental factors are associated with an elevated risk of CHD. Maternal diabetes mellitus (matDM) is associated with up to a fivefold increased risk of having an infant with CHD. The rising prevalence of diabetes mellitus has led to a growing interest in the use of experimental diabetic models to elucidate mechanisms underlying this associated risk for CHD. The purpose of this review is to provide a comprehensive summary of rodent models that are being used to investigate alterations in cardiac developmental pathways when exposed to a maternal diabetic setting and to summarize the key findings from these models. The majority of studies in the field have utilized the chemically induced model of matDM, but recent advances have also been made using diet based and genetic models. Each model provides an opportunity to investigate unique aspects of matDM and is invaluable for a comprehensive understanding of the molecular and cellular mechanisms underlying matDM-associated CHD.


Asunto(s)
Diabetes Gestacional/metabolismo , Cardiopatías Congénitas/etiología , Corazón/embriología , Hiperglucemia/metabolismo , Embarazo en Diabéticas/metabolismo , Animales , Diabetes Gestacional/genética , Femenino , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/genética , Embarazo , Embarazo en Diabéticas/genética
6.
Diabet Med ; 38(6): e14488, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33277730

RESUMEN

AIMS: To determine the fetal and maternal outcomes in pregnant women with Glucokinase-Maturity onset diabetes of the young (GCK-MODY). METHODS: We studied the obstetric and perinatal outcomes in 99 pregnancies of 34 women with GCK-MODY. The mutation status of the offspring was known in 29 and presumed in 33. Clinical outcomes were determined and compared between affected (n = 39) and unaffected (n = 23) offspring. RESULTS: 59% of pregnancies were treated with diet alone and 41% received insulin. Birthweight, percentage of large for gestational age (LGA) and caesarean section (CS) in GCK-unaffected offspring was significantly higher than in GCK-affected offspring (4.0 ± 0.7 vs. 3.4 ± 0.4 kg, p = 0.001), 15 (65%) vs. 5(13%) (p = 0.00006) and 17 (74%) vs. 11 (28%) (p = 0.001), respectively. We observed an earlier gestational age at delivery on insulin in unaffected offspring (38.3 ± 1.0 vs. 39.5 ± 1.5 weeks, p = 0.03) with no significant change in LGA (9 (82%) vs. 6 (50%); p = 0.12), and a higher rate of CS (8 [73%] vs. 3 [11%]; p < 0.001), and no change in small for gestational age (0 [0%] vs. 4 [14%]; p = 0.30) in affected offspring. CONCLUSION: Insulin therapy in unaffected offspring did not reduce LGA and was associated with earlier gestational age at delivery. Insulin treatment in GCK-affected offspring was associated with an increased incidence of CS, but did not adversely affect fetal outcome. Fetal genotype determines birthweight rather than treatment. Pre-pregnancy diagnosis of GCK-MODY, use of continuous glucose monitoring and non-invasive fetal genotyping may enable further investigation of targeted therapy in this condition.


Asunto(s)
ADN/genética , Diabetes Mellitus Tipo 2/genética , Glucoquinasa/genética , Mutación , Embarazo en Diabéticas/genética , Adulto , Peso al Nacer , Glucemia/metabolismo , Automonitorización de la Glucosa Sanguínea , Análisis Mutacional de ADN , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Femenino , Estudios de Seguimiento , Edad Gestacional , Glucoquinasa/metabolismo , Humanos , Incidencia , Linaje , Embarazo , Resultado del Embarazo , Embarazo en Diabéticas/sangre , Embarazo en Diabéticas/epidemiología , Estudios Retrospectivos , España/epidemiología
7.
Am J Obstet Gynecol ; 223(5): 753.e1-753.e14, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32416155

RESUMEN

BACKGROUND: Autophagy is highly active in neuroepithelial cells of the developing neuroepithelium, and impairment of autophagy leads to neural tube defects. In this study, we have found that maternal diabetes suppresses autophagy that leads to neural tube defects and consequent cellular imbalance in the endoplasmic reticulum where critical events occur, leading to the induction of diabetic embryopathy. Because the mammalian target of rapamycin pathway suppresses autophagy, we hypothesized that 70 kDa ribosomal protein S6 kinase 1 (p70S6K1), a major downstream effector of mammalian target of rapamycin, mediates the inhibitory effect of maternal diabetes on autophagy in the developing neuroepithelium. OBJECTIVE: We investigated whether p70S6K1 mediates the inhibitory effect of maternal diabetes on autophagy during neurulation. We also examined whether p70S6K1 deficiency restores autophagy and therefore relieves endoplasmic reticulum stress and inhibits maternal diabetes-induced apoptosis, which leads to reduction in neural tube defect incidence in diabetic embryopathy. STUDY DESIGN: Female p70S6K1 heterogeneous knockout (p70S6K1+/-) mice were bred with male p70S6K1 heterogeneous knockout (p70S6K1+/-) mice to generate wild-type (WT), p70S6K1+/- and p70S6K1 knockout (p70S6K1-/-) embryos. Embryos at embryonic day 8.5 were harvested for the assessment of indices of autophagy, endoplasmic reticulum stress, and apoptosis. Neural tube defect incidence in embryos was determined at embryonic day 10.5. For in vitro studies, small interfering RNA knockdown of p70S6K1 in C17.2 mouse neural stem cells was used to determine the effect of p70S6K1 deficiency on autophagy impairment and endoplasmic reticulum stress under high glucose conditions. RESULTS: Knockout of the Rps6kb1 gene, which encodes for p70S6K1, ameliorated maternal diabetes-induced NTDs and restored autophagosome formation in neuroepithelial cells suppressed by maternal diabetes. Maternal diabetes-suppressed conversion of LC3-I (microtubule-associated protein 1A/1B-light chain 3) to LC3-II, an index of autophagic activity, in neurulation stage embryos was abrogated in the absence of p70S6K1. p70S6K1 knockdown in neural stem cells also restored autophagosome formation and the conversion of LC3-I to LC3-II. The activation of the major unfolded protein response, indicated by phosphorylation of inositol-requiring enzyme 1 alpha, and protein kinase R-like endoplasmic reticulum kinase, and eukaryotic translation initiation factor 2α, and the increase of the endoplasmic reticulum stress marker, C/EBP homologous protein, were induced by maternal diabetes in vivo and high glucose in vitro. Unfolded protein response and endoplasmic reticulum stress induced by maternal diabetes or high glucose were reduced by Rps6kb1 deletion or p70S6K1 knockdown, respectively. Rps6kb1 knockout blocked maternal diabetes-induced caspase cleavage and neuroepithelial cell apoptosis. The superoxide dismutase mimetic Tempol abolished high glucose-induced p70S6K1 activation. CONCLUSION: The study revealed the critical involvement of p70S6K1 in the pathogenesis of diabetic embryopathy.


Asunto(s)
Autofagia/genética , Estrés del Retículo Endoplásmico/genética , Enfermedades Fetales/genética , Células-Madre Neurales/metabolismo , Defectos del Tubo Neural/genética , Embarazo en Diabéticas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Respuesta de Proteína Desplegada/genética , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Glucemia/metabolismo , Óxidos N-Cíclicos/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Enfermedades Fetales/etiología , Enfermedades Fetales/metabolismo , Glucosa/farmacología , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/metabolismo , Células Neuroepiteliales/efectos de los fármacos , Células Neuroepiteliales/metabolismo , Neurulación/genética , Estrés Oxidativo , Embarazo , Embarazo en Diabéticas/metabolismo , Marcadores de Spin , Respuesta de Proteína Desplegada/efectos de los fármacos
8.
Ceska Gynekol ; 85(2): 124-130, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32527107

RESUMEN

OBJECTIVE: The aim of this article is to present a summary of the actual diagnostic possibilities and differentiation of MODY (Maturity-Onset Diabetes of the Young) from gestational diabetes (GDM) found during routine screening, and specific aspects of care and treatment of MODY during pregnancy and early postpartum period. DESIGN: Rewiev. SETTINGS: Centre for Research of Diabetes, Metabolism and Nutrition; Second Department of Internal Medicine University Hospital Královské Vinohrady and Third Faculty of Medicine, Prague. Department of Internal Medicine, Second Faculty of Medicine, Charles University, Prague. METHODS: Recent publications selected in PubMed with the key words MODY, gestational diabetes. RESULTS: Many patients with MODY, especially the glucokinase MODY, can be first diagnosed during pregnancy. It is estimated that MODY patients account for up to 5% of GDM cases found in routine screening of GDM. MODY should be considered in lean women around 25 years of age, with a positive family history of diabetes in one of the parents. The differentiation of MODY from GDM is of particular importance not only for the different management and goals of antidiabetic therapy and planning ultrasound controls of fetal growth during pregnancy, but also because of the risk of hyperinsulinemic hypoglycemia in newborns. CONCLUSION: Recognition of MODY during pregnancy and adherence to existing recommendations concerning specific care of these patients is essential for the optimal course of their pregnancy and proper care of the newborn in the early postpartum period.


Asunto(s)
Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/diagnóstico , Glucoquinasa/genética , Embarazo en Diabéticas/diagnóstico , Embarazo en Diabéticas/genética , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Gestacional/tratamiento farmacológico , Diabetes Gestacional/genética , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Recién Nacido , Insulina/uso terapéutico , Mutación , Periodo Posparto , Embarazo , Resultado del Embarazo , Embarazo en Diabéticas/tratamiento farmacológico , Embarazo en Diabéticas/terapia , Resultado del Tratamiento
9.
Biochem Biophys Res Commun ; 508(4): 1149-1154, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30554659

RESUMEN

Diabetes in pregnancy constitutes an unfavorable environment for embryonic and fetal development, where the child has a higher risk of perinatal morbidity and mortality, with high incidence of congenital malformations and predisposition to long-term metabolic diseases that increase with a hypercaloric diet. To analyze whether hyperglycemia differentially affects proliferation, apoptosis, and mRNA expression in cells from children of normoglycemic pregnancies (NGPs) and diabetes mellitus pregnancies (DMPs), we used umbilical cord Wharton jelly cells as a research model. Proliferation assays were performed to analyze growth and determine the doubling time, and the rate of apoptosis was determined by flow cytometry-annexin-V assays. AMPK, BNIP3, HIF1α, and p53 mRNA gene expression was assessed by semi-quantitative RT-PCR. We found that hyperglycemia decreased proliferation in a statistically significant manner in NGP cells treated with 40 mM D-glucose and in DMP cells treated with 30 and 40 mM D-glucose. Apoptosis increased in hyperglycemic conditions in NGP and DMP cells. mRNA expression of BNIP3 and p53 was significantly increased in cells from DMPs but not in cells from NGPs. We found evidence that maternal irregular metabolic conditions, like diabetes with hyperglycemia in culture, affect biological properties of fetal cells. These observations could be a constituent of fetal programming.


Asunto(s)
Apoptosis/genética , Hiperglucemia/genética , Proteínas de la Membrana/genética , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/patología , Proteínas Proto-Oncogénicas/genética , Proteína p53 Supresora de Tumor/genética , Cordón Umbilical/patología , Gelatina de Wharton/metabolismo , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Proliferación Celular/genética , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/metabolismo , Embarazo , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
10.
Diabet Med ; 36(2): 252-255, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30362177

RESUMEN

BACKGROUND: Glucokinase-maturity-onset diabetes of the young (GCK-MODY) is a form of diabetes caused by heterozygous inactivating mutations in the GCK gene. Affected individuals maintain their fasting glucose levels at a higher set point (5.4-8.3 mmol/l) than the general population. Hyperglycaemia in women with Type 1 or Type 2 diabetes is known to confer increased risk of fetal congenital abnormalities. The association between GCK-MODY and congenital abnormalities, however, remains uncertain. CASE REPORT: A 35-year-old woman in her third pregnancy was diagnosed with gestational diabetes at 13 weeks' gestation (fasting blood glucose 6.0 mmol/L, 1-h blood glucose 9.2 mmol/l, 2-h blood glucose 7.3 mmol/l). The morphology scan at 19+2 weeks' gestation showed a Type III sacral agenesis. The woman elected to terminate the pregnancy. Her postpartum oral glucose tolerance test was suggestive of GCK-MODY (fasting blood glucose 7.4 mmol/l, 1-h blood glucose 9.3 mmol/l, 2-h blood glucose 7.3 mmol/l). Mutation analysis of the GCK gene identified a novel heterozygous GCK missense mutation, p.V199M, classified as likely pathogenic, providing molecular confirmation of the suspected GCK-MODY diagnosis. DISCUSSION: Sacral agenesis is a rare form of sacral abnormality affecting 0.005% to 0.1% of pregnancies. It is a subtype of the caudal regression sequence, a cardinal feature of diabetic embryopathy. This case raises the question as to whether hyperglycaemia in GCK-MODY may increase the risk of fetal caudal regression syndrome as reported in women with pre-existing diabetes mellitus. Improved diagnostic rates of GCK-MODY, and MODY registers that include pregnancy outcomes, are important to further elucidate risk of congenital abnormalities in GCK-MODY.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Feto/anomalías , Glucoquinasa/genética , Mutación Missense/genética , Embarazo en Diabéticas/genética , Sacro/anomalías , Adulto , Femenino , Heterocigoto , Humanos , Hiperglucemia/complicaciones , Embarazo , Factores de Riesgo
11.
Curr Diab Rep ; 18(3): 12, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29450745

RESUMEN

PURPOSE OF REVIEW: Monogenic diabetes accounts for 1-2% of all diabetes cases, but is frequently misdiagnosed as type 1, type 2, or gestational diabetes. Accurate genetic diagnosis directs management, such as no pharmacologic treatment for GCK-MODY, low-dose sulfonylureas for HNF1A-MODY and HNF4A-MODY, and high-dose sulfonylureas for KATP channel-related diabetes. While diabetes treatment is defined for the most common causes of monogenic diabetes, pregnancy poses a challenge to management. Here, we discuss the key issues in pregnancy affected by monogenic diabetes. RECENT FINDINGS: General recommendations for pregnancy affected by GCK-MODY determine need for maternal insulin treatment based on fetal mutation status. However, a recent study suggests macrosomia and miscarriage rates may be increased with this strategy. Recent demonstration of transplacental transfer of sulfonylureas also raises questions as to when insulin should be initiated in sulfonylurea-responsive forms of monogenic diabetes. Pregnancy represents a challenge in management of monogenic diabetes, where factors of maternal glycemic control, fetal mutation status, and transplacental transfer of medication must all be taken into consideration. Guidelines for pregnancy affected by monogenic diabetes will benefit from large, prospective studies to better define the need for and timing of initiation of insulin treatment.


Asunto(s)
Diabetes Mellitus Tipo 2/terapia , Embarazo en Diabéticas/terapia , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Glucoquinasa/genética , Factores Nucleares del Hepatocito/genética , Humanos , Mutación , Canales de Potasio/genética , Embarazo , Embarazo en Diabéticas/diagnóstico , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/fisiopatología
12.
Am J Perinatol ; 33(13): 1319-1326, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27571483

RESUMEN

The genetic architecture of diabetes mellitus in general and in pregnancy is complex, owing to the multiple types of diabetes that comprise both complex/polygenic forms and monogenic (largely caused by a mutation in a single gene) forms such as maturity-onset diabetes of the young (MODY). Type 1 diabetes (T1D) and type 2 diabetes (T2D) have complex genetic etiologies, with over 40 and 90 genes/loci, respectively, implicated that interact with environmental/lifestyle factors. The genetic etiology of gestational diabetes mellitus has largely been found to overlap that of T2D. Genetic testing for complex forms of diabetes is not currently useful clinically, but genetic testing for monogenic forms, particularly MODY, has important utility for determining treatment, managing risk in family members, and pregnancy management. In particular, diagnosing MODY2, caused by GCK mutations, indicates that insulin should not be used, including during pregnancy, with the possible exception of an unaffected pregnancy during the third trimester to prevent macrosomia. A relatively simple method for identifying women with MODY2 has been piloted. MODY1, caused by HNF4A mutations, can paradoxically cause neonatal hyperinsulinemic hypoglycemia and macrosomia, indicating that detecting these cases is also clinically important. Diagnosing all MODY types provides opportunities for diagnosing other family members.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/genética , Embarazo en Diabéticas/genética , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 2/diagnóstico , Femenino , Glucoquinasa/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Humanos , Embarazo
13.
Am J Physiol Endocrinol Metab ; 309(5): E487-99, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26173459

RESUMEN

Maternal diabetes in mice induces heart defects similar to those observed in human diabetic pregnancies. Diabetes enhances apoptosis and suppresses cell proliferation in the developing heart, yet the underlying mechanism remains elusive. Apoptosis signal-regulating kinase 1 (ASK1) activates the proapoptotic c-Jun NH2-terminal kinase 1/2 (JNK1/2) leading to apoptosis, suggesting a possible role of ASK1 in diabetes-induced heart defects. We aimed to investigate whether ASK1 is activated in the heart and whether deleting the Ask1 gene blocks diabetes-induced adverse events and heart defect formation. The ASK1-JNK1/2 pathway was activated by diabetes. Deleting Ask1 gene significantly reduced the rate of heart defects, including ventricular septal defects (VSDs) and persistent truncus arteriosus (PTA). Additionally, Ask1 deletion diminished diabetes-induced JNK1/2 phosphorylation and its downstream transcription factors and endoplasmic reticulum (ER) stress markers. Consistent with this, caspase activation and apoptosis were blunted. Ask1 deletion blocked the increase in cell cycle inhibitors (p21 and p27) and the decrease in cyclin D1 and D3 and reversed diabetes-repressed cell proliferation. Ask1 deletion also restored the expression of BMP4, NKX2.5, and GATA5, Smad1/5/8 phosphorylation, whose mutations or deletion result in reduced cell proliferation, VSD, and PTA formation. We conclude that ASK1 may mediate the teratogenicity of diabetes through activating the JNK1/2-ER stress pathway and inhibiting cell cycle progression, thereby impeding the cardiogenesis pathways essential for ventricular septation and outflow tract development.


Asunto(s)
Apoptosis/genética , Estrés del Retículo Endoplásmico/genética , Defectos del Tabique Interventricular/genética , Corazón/embriología , MAP Quinasa Quinasa Quinasa 5/genética , Embarazo en Diabéticas/genética , Teratogénesis/genética , Tronco Arterial Persistente/genética , Animales , Proteína Morfogenética Ósea 4/metabolismo , Proliferación Celular , Ciclina D1/metabolismo , Ciclina D3/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Factor de Transcripción GATA5/metabolismo , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interventricular/etiología , Defectos del Tabique Interventricular/metabolismo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Fosforilación , Embarazo , Embarazo en Diabéticas/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Factores de Transcripción/metabolismo , Tronco Arterial Persistente/etiología , Tronco Arterial Persistente/metabolismo
14.
Diabet Med ; 32(12): 1596-601, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26043405

RESUMEN

AIM: To determine if the previously published clinical criteria for identifying glucokinase monogenic diabetes [GCK gene mutation in maturity-onset diabetes of the young (GCK-MODY)], an elevated antenatal fasting blood glucose of 5.5-8.0 mmol/l, an increment of < 4.6 mmol/l at 2 h in an oral glucose tolerance test and slim are applicable in a large multi-ethnic cohort of women with gestational diabetes. METHODS: We analysed de-identified data from all women with gestational diabetes, diagnosed using the Australasian Diabetes in Pregnancy Society (1998) Australian criteria at our institution between 1993 and 2013, making comparisons among those with complete antenatal data including: diagnostic oral glucose tolerance test results meeting the above criteria; pregestational BMI; birth outcomes; and postpartum oral glucose tolerance test data. We categorized these women into two groups: Group A1 had a BMI ≤ 21 kg/m(2) and Group A2 had a BMI > 21 kg/m(2) and < 25 kg/m(2). RESULTS: Of the 302 women meeting the study entry criteria, we had complete data including a postpartum oral glucose tolerance test result for 171 women: 54 in Group A1 and 117 in Group A2. Ethnicity was significantly different between the groups. The oral glucose tolerance test and postpartum HbA1c results identified few women ( < 14%) in Group A1 and Group A2 who still had 'possible GCK-MODY'. CONCLUSIONS: Our findings indicate that previously recommended clinical criteria for the identification of women likely to have GCK-MODY lack specificity in a cohort of women with multi-ethnic backgrounds. Using these criteria to select women for testing for GCK-MODY in pregnancy would therefore be costly and is likely to yield few women positive for this condition.


Asunto(s)
Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Gestacional/diagnóstico , Glucoquinasa/genética , Mutación , Embarazo en Diabéticas/diagnóstico , Diagnóstico Prenatal , Adulto , Glucemia/análisis , Índice de Masa Corporal , Estudios de Cohortes , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/sangre , Diabetes Gestacional/enzimología , Diabetes Gestacional/genética , Diagnóstico Diferencial , Registros Electrónicos de Salud , Femenino , Estudios de Seguimiento , Glucoquinasa/metabolismo , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Humanos , Nueva Gales del Sur , Embarazo , Embarazo en Diabéticas/sangre , Embarazo en Diabéticas/enzimología , Embarazo en Diabéticas/genética , Estudios Retrospectivos
15.
Biol Reprod ; 90(6): 139, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24829025

RESUMEN

Offspring of diabetic mothers are susceptible to the onset of metabolic syndromes, such as type 2 diabetes and obesity at adulthood, and this trend can be inherited between generations. Genetics cannot fully explain how the noncommunicable disease in offspring of diabetic mothers is caused and inherited by the next generations. Many studies have confirmed that epigenetics may be crucial for the detrimental effects on offspring exposed to the hyperglycemic environment. Although the adverse effects on epigenetics in offspring of diabetic mothers may be the result of the poor intrauterine environment, epigenetic modifications in oocytes of diabetic mothers are also affected. Therefore, the present review is focused on the epigenetic alterations in oocytes and embryos of diabetic mothers. Furthermore, we also discuss initial mechanistic insight on maternal diabetes mellitus causing alterations of epigenetic modifications.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Epigénesis Genética/fisiología , Embarazo en Diabéticas/genética , Efectos Tardíos de la Exposición Prenatal/genética , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Humanos , Lactante , Obesidad/complicaciones , Obesidad/genética , Obesidad/fisiopatología , Embarazo , Embarazo en Diabéticas/etiología , Embarazo en Diabéticas/fisiopatología , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología
16.
Artículo en Inglés | MEDLINE | ID: mdl-24561380

RESUMEN

Pregestational diabetes mellitus (type 1 and type 2) affects about 1% of the obstetric population. In diabetes, persistent hyperglycemia can be a source of DNA damage via overproduction of reactive oxygen species (ROS). Using the cytokinesis-block micronucleus (CBMN) test, we measured the frequencies of micronuclei (MN) per 1000 binucleated (BN) cells in pregnant women (mothers) with type 1 diabetes mellitus (T1DM) and in their newborns. Peripheral blood lymphocytes were collected from 17 pregnant women with T1DM and cord-blood lymphocytes from their 17 newborns. The control group included 40 pregnant women (mothers) without diabetes mellitus (DM) and their 40 newborns. In the group of pregnant women with T1DM, the mean number of MN per 1000 BN cells was 2.35 (±1.07), significantly (p<0.001) higher than in the control group of pregnant women (0.86±0.90). The frequency value in the group of newborns of T1DM mothers was 1.42 (±0.60), significantly (p<0.05) higher than in the corresponding control group (0.67±0.79). The value in the group of mothers with T1DM was significantly (p<0.05) higher than in their newborns. Comparing mothers without DM with their newborns, no significant frequency differences were observed. No significant correlations were observed between MN frequencies in mothers with T1DM and either the frequencies in their newborns, the duration of diabetes, or HbA1C levels. Our results indicate that T1DM is accompanied by increased frequencies of MN in pregnant women and their newborns.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Recién Nacido , Micronúcleos con Defecto Cromosómico/inducido químicamente , Micronúcleos con Defecto Cromosómico/embriología , Embarazo en Diabéticas/genética , Estudios de Casos y Controles , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Recién Nacido/sangre , Linfocitos/patología , Micronúcleos con Defecto Cromosómico/estadística & datos numéricos , Pruebas de Micronúcleos , Estrés Oxidativo/genética , Embarazo , Embarazo en Diabéticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
17.
Ginekol Pol ; 85(1): 18-25, 2014 Jan.
Artículo en Polaco | MEDLINE | ID: mdl-24505959

RESUMEN

AIM: to study the frequency of genetic variants of eNOS and ACE polimorphism and their possible influence on the course of diabetic pregnancy and perinatal outcome. MATERIAL AND METHODS: 107 pregnant women with type 1 diabetes, treated at the Department of Obstetrics and Women's Diseases between 2008-2011, were enrolled into the study. Ninety six (90%) of the patients delivered at term. All women were treated with intensive insulin therapy Glucose control was performed by means of self-monitoring with glucometers. The target fasting glucose levels were below 90 mg/dl (5.0 mmol/l) and postprandial below 120 mg/dl (6.7 mmol/I). DNA for the analysis of polimorphisms was extracted from the leukocytes. Afterwards, the number of specific eNOS and ACE genotypes was calculated and the subgroups of alleles of these two genes were created. RESULTS: Subjects with heterozygote genotype eNOS GT and ACE ID constituted the largest group of patients (24/22%); the smallest group presented eNOS TT (ACE II, ID, DD) genotype (8/8% of the whole studied group). Next, selected genotypes were analyzed in relation to the metabolic status, duration of diabetes and perinatal outcome. RESULTS: Our results enabled us to conclude that, despite identical treatment of all gravidas, diabetic patients with eNOS TT polimorphism presented with the highest body weight, and the strongest lipid and glucose disturbances, what probably resulted in marosomic neonatal weight. CONCLUSIONS: The eNOS and ACE genetic variants may affect the course of a diabetic pregnancy in terms of metabolic control and perinatal outcome.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Genotipo , Óxido Nítrico Sintasa de Tipo III/genética , Peptidil-Dipeptidasa A/genética , Polimorfismo de Nucleótido Simple , Embarazo en Diabéticas/genética , Adulto , Alelos , Diabetes Mellitus Tipo 1/complicaciones , Femenino , Humanos , Embarazo , Resultado del Embarazo , Análisis de Regresión , Adulto Joven
18.
Gene ; 922: 148557, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38740354

RESUMEN

The primary aim of this study was to explore the impact of diabetes on matrix metalloproteases and tissue inhibitors, crucial factors for successful implantation, and to elucidate the molecular mechanisms that undergo changes in the endometrium and the embryo during diabetic pregnancies. In this investigation, we established a streptozotocin-induced diabetic pregnant rat model. Microarray analysis followed by RT-PCR was utilized to identify gene regions exhibiting expression alterations. Subsequently, we assessed the effects of MMPs and tissue inhibitors using ELISA and immunohistochemistry techniques, in addition to analyzing changes at the genetic level. Diabetes led to the upregulation of MMP3, MMP9, and MMP20 on the 6.5th day of pregnancy, while causing the downregulation of MMP3, MMP9, and MMP11 on the 8.5th day of pregnancy. TIMP1 expression was downregulated on the 8.5th day compared to the control group. No statistically significant differences were observed between the groups regarding other TIMP expressions. KEGG pathway analysis revealed that diabetes induced alterations in the expression of genes associated with certain microRNAs, as well as signaling pathways such as cAMP, calcium, BMP, p53, MAPK, PI3K-Akt, Jak-STAT, Hippo, Wnt, and TNF. Additionally, gene ontology analysis unveiled changes in membrane structures, extracellular matrix, signaling pathways, ion binding, protein binding, cell adhesion molecule binding, and receptor-ligand activity. This study serves as a valuable guide for investigating the mechanisms responsible for complications in diabetic pregnancies. By revealing the early-stage effects of diabetes, it offers insight into the development of new diagnostic and treatment approaches, ultimately contributing to improved patient care.


Asunto(s)
Diabetes Mellitus Experimental , Endometrio , Animales , Femenino , Embarazo , Endometrio/metabolismo , Ratas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Transducción de Señal , Embrión de Mamíferos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Embarazo en Diabéticas/metabolismo , Embarazo en Diabéticas/genética , Implantación del Embrión/genética , Ratas Sprague-Dawley , MicroARNs/genética , MicroARNs/metabolismo
19.
J Mol Cell Cardiol ; 60: 129-41, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23619295

RESUMEN

Cardiovascular malformations are the most common manifestation of diabetic embryopathy. The molecular mechanisms underlying the teratogenic effect of maternal diabetes have not been fully elucidated. Using genome-wide expression profiling, we previously demonstrated that exposure to maternal diabetes resulted in dysregulation of the hypoxia-inducible factor 1 (HIF-1) pathway in the developing embryo. We thus considered a possible link between HIF-1-regulated pathways and the development of congenital malformations. HIF-1α heterozygous-null (Hif1a(+/-)) and wild type (Wt) littermate embryos were exposed to the intrauterine environment of a diabetic mother to analyze the frequency and morphology of congenital defects, and assess gene expression changes in Wt and Hif1a(+/-) embryos. We observed a decreased number of embryos per litter and an increased incidence of heart malformations, including atrioventricular septal defects and reduced myocardial mass, in diabetes-exposed Hif1a(+/-) embryos as compared to Wt embryos. We also detected significant differences in the expression of key cardiac transcription factors, including Nkx2.5, Tbx5, and Mef2C, in diabetes-exposed Hif1a(+/-) embryonic hearts compared to Wt littermates. Thus, partial global HIF-1α deficiency alters gene expression in the developing heart and increases susceptibility to congenital defects in a mouse model of diabetic pregnancy.


Asunto(s)
Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Corazón/embriología , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas Musculares/biosíntesis , Embarazo en Diabéticas/metabolismo , Animales , Embrión de Mamíferos/patología , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Ratones , Ratones Mutantes , Proteínas Musculares/genética , Embarazo , Embarazo en Diabéticas/genética , Embarazo en Diabéticas/patología
20.
Reprod Biol Endocrinol ; 11: 119, 2013 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-24378208

RESUMEN

BACKGROUND: Maternal diabetes mellitus not only has severe deleterious effects on fetal development, but also it affects transmission to the next generation. However, the underlying mechanisms for these effects are still not clear. METHODS: We investigated the methylation patterns and expressions of the imprinted genes Peg3, Snrpn, and H19 in mid-gestational placental tissues and on the whole fetus utilizing the streptozotocin (STZ)-induced hyperglycemic mouse model for quantitative analysis of methylation by PCR and quantitative real-time PCR. The protein expression of Peg3 was evaluated by Western blot. RESULTS: We found that the expression of H19 was significantly increased, while the expression of Peg3 was significantly decreased in dpc10.5 placentas of diabetic mice. We further found that the methylation level of Peg3 was increased and that of H19 was reduced in dpc10.5 placentas of diabetic mice. When pronuclear embryos of normal females were transferred to normal/diabetic (NN/ND) pseudopregnant females, the methylation and expression of Peg3 in placentas was also clearly altered in the ND group compared to the NN group. However, when the pronuclear embryos of diabetic female were transferred to normal pesudopregnant female mice (DN), the methylation and expression of Peg3 and H19 in dpc10.5 placentas was similar between the two groups. CONCLUSIONS: We suggest that the effects of maternal diabetes on imprinted genes may primarily be caused by the adverse uterus environment.


Asunto(s)
Diabetes Mellitus/genética , Desarrollo Fetal/genética , Impresión Genómica , Embarazo en Diabéticas/genética , Útero/metabolismo , Animales , Western Blotting , Metilación de ADN , Femenino , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Placenta/metabolismo , Embarazo , Embarazo en Diabéticas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Nucleares snRNP/genética , Proteínas Nucleares snRNP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA