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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mov Disord ; 37(2): 365-374, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34820905

RESUMEN

BACKGROUND: The dystonias are a heterogeneous group of hyperkinetic disorders characterized by sustained or intermittent muscle contractions that cause abnormal movements and/or postures. Although more than 200 causal genes are known, many cases of primary dystonia have no clear genetic cause. OBJECTIVES: To identify the causal gene in a consanguineous family with three siblings affected by a complex persistent generalized dystonia, generalized epilepsy, and mild intellectual disability. METHODS: We performed exome sequencing in the parents and two affected siblings and characterized the expression of the identified gene by immunohistochemistry in control human and zebrafish brains. RESULTS: We identified a novel missense variant (c.142G>A (NM_032192); p.Glu48Lys) in the protein phosphatase 1 regulatory inhibitor subunit 1B gene (PPP1R1B) that was homozygous in all three siblings and heterozygous in the parents. This gene is also known as dopamine and cAMP-regulated neuronal phosphoprotein 32 (DARPP-32) and has been involved in the pathophysiology of abnormal movements. The uncovered variant is absent in public databases and modifies the conserved glutamate 48 localized close to the serine 45 phosphorylation site. The PPP1R1B protein was shown to be expressed in cells and regions involved in movement control, including projection neurons of the caudate-putamen, substantia nigra neuropil, and cerebellar Purkinje cells. The latter cells were also confirmed to be positive for PPP1R1B expression in the zebrafish brain. CONCLUSIONS: We report the association of a PPP1R1B/DARPP-32 variant with generalized dystonia in man. It might be relevant to include the sequencing of this new gene in the diagnosis of patients with otherwise unexplained movement disorders. © 2021 International Parkinson and Movement Disorder Society.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Distonía , Trastornos Distónicos , Animales , Trastornos Distónicos/genética , Homocigoto , Humanos , Pez Cebra
2.
Pediatr Res ; 82(5): 741-748, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28678770

RESUMEN

BackgroundLactoferrin (LTF) could play a beneficial role in insulin resistance and diabetes, but the association of its gene variants with cardio-metabolic disorders in children has not been investigated. This study aimed to examine the relationship between LTF variants, plasma LTF concentrations, and cardio-metabolic risk factors in French-Canadian children.MethodsThe study cohort comprises 1,749 French Canadians aged 9, 13, and 16 years. The association of 13 LTF polymorphisms, metabolic parameters, and plasma LTF levels was tested in this cross-sectional, province-wide school-based survey.ResultsNone of the genetic association remained significant after correction for multiple testing and LTF SNPs were not associated with LTF levels. Plasma LTF was positively correlated with body mass index (r2=0.2245, P=0.0011) and weight (r2=0.2515, P=0.0008). After segregating according to high-density lipoprotein cholesterol (HDL-C), the association remained only in subjects exhibiting low HDL-C (r2=0.3868, P=0.0002 for body mass index and r2=0.3665, P=0.0004 for weight). In girls, plasma LTF was positively correlated with total cholesterol (r2=0.2231, P=0.0378), LDL cholesterol (r2=0.2409, P=0.0246), and apolipoprotein B (r2=0.2478, P=0.0207).ConclusionsWe found no association between LTF gene variants and metabolic parameters following correction for multiple testing. HDL-C and gender-specific positive associations were evidenced between plasma LTF, anthropometric profile, and lipid levels.


Asunto(s)
Lactoferrina/sangre , Lactoferrina/genética , Síndrome Metabólico/sangre , Síndrome Metabólico/genética , Polimorfismo de Nucleótido Simple , Adolescente , Factores de Edad , Índice de Masa Corporal , Peso Corporal , Niño , Estudios Transversales , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Encuestas Epidemiológicas , Humanos , Lípidos/sangre , Masculino , Síndrome Metabólico/diagnóstico , Síndrome Metabólico/epidemiología , Fenotipo , Quebec/epidemiología , Factores de Riesgo , Factores Sexuales
3.
BMC Gastroenterol ; 15: 119, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26376914

RESUMEN

BACKGROUND: Metabolic alterations relevant to postprandial dyslipidemia were previously identified in the intestine of obese insulin-resistant subjects. The aim of the study was to identify the genes deregulated by systemic insulin resistance in the intestine of severely obese subjects. METHODS: Transcripts from duodenal samples of insulin-sensitive (HOMA-IR < 3, n = 9) and insulin-resistant (HOMA-IR > 7, n = 9) obese subjects were assayed by microarray (Illumina HumanHT-12). RESULTS: A total of 195 annotated genes were identified as differentially expressed between these two groups (Fold change > 1.2). Of these genes, 36 were found to be directly involved in known intestinal functions, including digestion, extracellular matrix, endocrine system, immunity and cholesterol metabolism. Interestingly, all differentially expressed genes (n = 8) implicated in inflammation and oxidative stress were found to be upregulated in the intestine of insulin-resistant compared to insulin-sensitive subjects. Metabolic pathway analysis revealed that several signaling pathways involved in immunity and inflammation were significantly enriched in differently expressed genes and were predicted to be activated in the intestine of insulin-resistant subjects. Using stringent criteria (Fold change > 1.5; FDR < 0.05), three genes were found to be significantly and differently expressed in the intestine of insulin-resistant compared to insulin-sensitive subjects: the transcripts of the insulinotropic glucose-dependant peptide (GIP) and of the ß-microseminoprotein (MSMB) were significantly reduced, but that of the humanin like-1 (MTRNR2L1) was significantly increased. CONCLUSION: These results underline that systemic insulin resistance is associated with remodeling of key intestinal functions. Moreover, these data indicate that small intestine metabolic dysfunction is accompanied with a local amplification of low-grade inflammatory process implicating several pathways. Genes identified in this study are potentially triggered throughout the development of intestinal metabolic abnormalities, which could contribute to dyslipidemia, a component of metabolic syndrome and diabetes.


Asunto(s)
Expresión Génica , Inflamación/genética , Resistencia a la Insulina/genética , Obesidad/genética , Obesidad/fisiopatología , ARN Mensajero/metabolismo , Adulto , Células CACO-2 , Duodeno , Femenino , Polipéptido Inhibidor Gástrico/genética , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Análisis por Micromatrices , Obesidad/patología , Estrés Oxidativo/genética , Proteínas de Secreción Prostática/genética , Proteínas/genética , Transducción de Señal/genética
4.
Am J Physiol Endocrinol Metab ; 304(1): E14-22, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23092912

RESUMEN

Low birth weight is associated with an increased risk for developing type 2 diabetes and metabolic diseases. The placental capacity to supply nutrients and oxygen to the fetus represents the main determiner of fetal growth. However, few studies have investigated the effects of maternal diet on the placenta. We explored placental adaptive proteomic processes implicated in response to maternal undernutrition. Rat term placentas from 70% food-restricted (FR30) mothers were used for a proteomic screen. Placental mitochondrial functions were evaluated using molecular and functional approaches, and ATP production was measured. FR30 drastically reduced placental and fetal weights. FR30 placentas displayed 14 proteins that were differentially expressed, including several mitochondrial proteins. FR30 induced a marked increase in placental mtDNA content and changes in mitochondrial functions, including modulation of the expression of genes implicated in biogenesis and bioenergetic pathways. FR30 mitochondria showed higher oxygen consumption but failed to maintain their ATP production. Maternal undernutrition induces placental mitochondrial abnormalities. Although an increase in biogenesis and bioenergetic efficiency was noted, placental ATP level was reduced. Our data suggest that placental mitochondrial defects may be implicated in fetoplacental pathologies.


Asunto(s)
Restricción Calórica/efectos adversos , Metabolismo Energético/fisiología , Retardo del Crecimiento Fetal/etiología , Fenómenos Fisiologicos Nutricionales Maternos , Mitocondrias/fisiología , Placenta/metabolismo , Animales , Eficiencia/fisiología , Femenino , Retardo del Crecimiento Fetal/metabolismo , Masculino , Intercambio Materno-Fetal/fisiología , Mitocondrias/metabolismo , Placenta/fisiología , Placenta/ultraestructura , Circulación Placentaria/fisiología , Embarazo , Ratas , Ratas Wistar
5.
Artículo en Inglés | MEDLINE | ID: mdl-37160315

RESUMEN

Following chemotherapy, a mediastinal germ cell tumor can lead to a mature teratoma that is composed of tissues derived from all three germ layers. Although teratoma is usually curable, in rare cases it can give rise to various somatic tumors and exceptionally it undergoes melanocytic neuroectodermal tumor (MNT) transformation, a process that is not well-described. We report a patient with a postchemotherapy thymic teratoma associated with an MNT component who, 10 years later, additionally presented a vertebral metastasis corresponding to an anaplastic MNT. Using exome sequencing of the mature teratoma, the MNT and its metastatic vertebral anaplastic MNT components, we identified 19 somatic mutations shared by at least two components. Six mutations were common to all three components, and three of them were located in the known cancer-related genes KRAS (p.E63K), TP53 (p.P222X), and POLQ (p.S447P). Gene set enrichment analysis revealed that the melanoma tumorigenesis pathway was enriched in mutated genes including the four major driver genes KRAS, TP53, ERBB4, and KDR, indicating that these genes may be involved in the development of the anaplastic MNT transformation of the teratoma. To our knowledge, this is the first molecular study realized on MNT. Understanding the clinicopathological and molecular characteristics of these tumors is essential to better understand their development and to improve therapeutics.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Tumores Neuroectodérmicos , Teratoma , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Teratoma/genética , Genómica
6.
Sci Transl Med ; 14(628): eabj7521, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34698500

RESUMEN

The drivers of critical coronavirus disease 2019 (COVID-19) remain unknown. Given major confounding factors such as age and comorbidities, true mediators of this condition have remained elusive. We used a multi-omics analysis combined with artificial intelligence in a young patient cohort where major comorbidities were excluded at the onset. The cohort included 47 "critical" (in the intensive care unit under mechanical ventilation) and 25 "non-critical" (in a non-critical care ward) patients with COVID-19 and 22 healthy individuals. The analyses included whole-genome sequencing, whole-blood RNA sequencing, plasma and blood mononuclear cell proteomics, cytokine profiling, and high-throughput immunophenotyping. An ensemble of machine learning, deep learning, quantum annealing, and structural causal modeling were used. Patients with critical COVID-19 were characterized by exacerbated inflammation, perturbed lymphoid and myeloid compartments, increased coagulation, and viral cell biology. Among differentially expressed genes, we observed up-regulation of the metalloprotease ADAM9. This gene signature was validated in a second independent cohort of 81 critical and 73 recovered patients with COVID-19 and was further confirmed at the transcriptional and protein level and by proteolytic activity. Ex vivo ADAM9 inhibition decreased severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uptake and replication in human lung epithelial cells. In conclusion, within a young, otherwise healthy, cohort of individuals with COVID-19, we provide the landscape of biological perturbations in vivo where a unique gene signature differentiated critical from non-critical patients. We further identified ADAM9 as a driver of disease severity and a candidate therapeutic target.


Asunto(s)
COVID-19 , Proteínas ADAM , Inteligencia Artificial , Humanos , Unidades de Cuidados Intensivos , Proteínas de la Membrana , Respiración Artificial , SARS-CoV-2
7.
Am J Physiol Endocrinol Metab ; 301(3): E548-59, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21712534

RESUMEN

Several studies have shown that maternal undernutrition leading to low birth weight predisposes offspring to the development of metabolic pathologies such as obesity. Using a model of prenatal maternal 70% food restriction diet (FR30) in rat, we evaluated whether postweaning high-fat (HF) diet would amplify the phenotype observed under standard diet. We investigated biological parameters as well as gene expression profile focusing on white adipose tissues (WAT) of adult offspring. FR30 procedure does not worsen the metabolic syndrome features induced by HF diet. However, FR30HF rats displayed catch-up growth to match the body weight of adult control HF animals, suggesting an increase of adiposity while showing hyperleptinemia and a blunted increase of corticosterone. Using quantitative RT-PCR array, we demonstrated that FR30HF rats exhibited leptin and Ob-Rb as well as many peptide precursor and receptor gene expression variations in WAT. We also showed that the expression of genes involved in adipogenesis was modified in FR30HF animals in a depot-specific manner. We observed an opposite variation of STAT3 phosphorylation levels, suggesting that leptin sensitivity is modified in WAT adult FR30 offspring. We demonstrated that 11ß-HSD1, 11ß-HSD2, GR, and MR genes are coexpressed in WAT and that FR30 procedure modifies gene expression levels, especially under HF diet. In particular, level variation of 11ß-HSD2, whose protein expression was detected by Western blotting, may represent a novel mechanism that may affect WAT glucocorticoid sensitivity. Data suggest that maternal undernutrition differently programs the adult offspring WAT gene expression profile that may predispose for altered fat deposition.


Asunto(s)
Tejido Adiposo/metabolismo , Grasas de la Dieta/metabolismo , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Adiposidad/efectos de los fármacos , Adiposidad/genética , Animales , Peso Corporal/genética , Femenino , Expresión Génica , Leptina/genética , Leptina/metabolismo , Masculino , Desnutrición/genética , Obesidad/genética , Obesidad/metabolismo , Fosforilación , Ratas , Ratas Wistar , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
8.
Hum Pathol ; 107: 39-45, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161030

RESUMEN

The clinical spectrum of coronavirus disease 2019 is getting wider with the exponential increase of patients worldwide. Initially described with flu-like symptoms, variable cutaneous manifestations have been reported, with only few histopathological descriptions. Detection of the virus in cutaneous samples has been assessed in very few cases until now, and the causative role of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has not been proven for every type of cutaneous manifestations yet. We aimed to describe histological features of cutaneous eruptions occurring concomitantly to SARS-CoV-2 infection and assess by immunochemistry and in situ hybridization using RNAscope validation techniques the presence of the virus in skin lesions. We retrieved all skin biopsies received in the departments of pathology and dermatopathology, University Hospital of Strasbourg, performed in hospitalized SARS-CoV-2-infected patients presenting concomitant cutaneous manifestations since March 2020. In situ hybridization and immunostaining using a polyclonal SARS nucleocapsid protein antibody were performed on each sample. Skin biopsies from six patients presenting morbilliform eruption concomitant to SARS-CoV-2 infection were available for evaluation. All six samples showed varying degrees of spongiosis, perivascular inflammatory infiltrates of the dermis, and, for some of them, discrete interface dermatitis. In situ hybridization and immunohistochemistry were negative in all cutaneous samples. Morbilliform rash concomitant to SARS-CoV-2 infection is characterized by mild and unspecific histopathological features with no detectable viral RNA and protein and appears then not to be directly caused by the virus. Even if, at least for a few cases, the differential diagnosis with drug hypersensitivity reaction can be difficult, these cutaneous eruptions seem to rather correspond to paraviral rashes.


Asunto(s)
COVID-19/complicaciones , Dermatitis/virología , SARS-CoV-2/patogenicidad , Enfermedades de la Piel/virología , Dermatitis/patología , Femenino , Humanos , Inmunohistoquímica/métodos , Masculino , ARN Viral/genética , Piel/patología , Piel/virología , Enfermedades de la Piel/patología
11.
Antioxid Redox Signal ; 24(14): 813-36, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-26981846

RESUMEN

SIGNIFICANCE: Lactoferrin (Lf) is a nonheme iron-binding glycoprotein strongly expressed in human and bovine milk and it plays many functions during infancy such as iron homeostasis and defense against microorganisms. In humans, Lf is mainly expressed in mucosal epithelial and immune cells. Growing evidence suggests multiple physiological roles for Lf after weaning. RECENT ADVANCES: The aim of this review is to highlight the recent advances concerning multifunctional Lf activities. CRITICAL ISSUES: First, we will provide an overview of the mechanisms related to Lf intrinsic synthesis or intestinal absorption as well as its interaction with a wide spectrum of mammalian receptors and distribution in organs and cell types. Second, we will discuss the large variety of its physiological functions such as iron homeostasis, transportation, immune regulation, oxidative stress, inflammation, and apoptosis while specifying the mechanisms of action. Third, we will focus on its recent physiopathology implication in metabolic disorders, including obesity, type 2 diabetes, and cardiovascular diseases. Additional efforts are necessary before suggesting the potential use of Lf as a diagnostic marker or as a therapeutic tool. FUTURE DIRECTIONS: The main sources of Lf in human cardiometabolic disorders should be clarified to identify new perspectives for future research and develop new strategies using Lf in therapeutics. Antioxid. Redox Signal. 24, 813-836.


Asunto(s)
Lactoferrina/fisiología , Enfermedades Metabólicas/metabolismo , Animales , Humanos , Factores Inmunológicos/fisiología , Hierro/metabolismo , Estrés Oxidativo , Unión Proteica , Transducción de Señal
12.
PLoS One ; 11(11): e0166138, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27902700

RESUMEN

CONTEXT: Lactoferrin (Lf) is an important protein found on mucosal surfaces, within neutrophils and various cells, and in biological fluids. It displays multiple functions, including iron-binding as well as antimicrobial, immunomodulatory and anti-inflammatory activities. Although Lf ingestion has been suggested to cause adiposity reduction in murine models and humans, its relationship with insulin resistance (IR) has not been studied thoroughly. OBJECTIVE: To establish the association between circulating Lf levels, glucose status and blood lipid/lipoprotein profile. METHODS: Two independent cohorts were examined: lean to moderately obese women admitted for gynecological surgery (n = 53) and severely obese subjects undergoing biliopancreatic diversion (n = 62). RESULTS: Although body mass index (BMI) and total body fat mass were negatively associated with Lf, IR (assessed by the HOMA-IR index) was positively and independently associated with plasma Lf concentrations of the first cohort of lean to moderately obese women. These observations were validated in the second cohort in view of the positive correlation between plasma Lf concentrations and the HOMA-IR index, but without a significant association with the body mass index (BMI) of severely obese subjects. In subsamples of severely obese subjects matched for sex, age and BMI, but with either relatively low (1.89 ± 0.73) or high (13.77 ± 8.81) IR states (according to HOMA-IR), higher plasma Lf levels were noted in insulin-resistant vs insulin-sensitive subjects (P<0.05). Finally, Lf levels were significantly higher in lean to moderately obese women than in severely obese subjects (P<0.05). CONCLUSION: Our findings revealed that plasma Lf levels are strongly associated with IR independently of total adiposity, which suggests an intriguing Lf regulation mechanism in conditions of obesity and IR.


Asunto(s)
Adiposidad/fisiología , Biomarcadores/sangre , Resistencia a la Insulina , Lactoferrina/sangre , Obesidad/fisiopatología , Delgadez/fisiopatología , Adulto , Glucemia/metabolismo , Estudios de Cohortes , Femenino , Humanos , Lípidos/sangre , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad
13.
Diabetes ; 65(3): 554-60, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631739

RESUMEN

The adequate control of glucose homeostasis during both gestation and early postnatal life is crucial for the development of the fetoplacental unit and adaptive physiological responses at birth. Growing evidences indicate that apelin and its receptor, APJ, which are expressed across a wide range of tissues, exert important roles in glucose homeostasis in adults. However, little is known about the function of the apelinergic system during gestation. In this study, we evaluated the activity of this system in rats, the role of apelin in fetal and neonatal glucose homeostasis, and its modulation by maternal food restriction. We found that 1) the apelinergic system was expressed at the fetoplacental interface and in numerous fetal tissues, 2) ex vivo, the placenta released high amounts of apelin in late gestation, 3) intravenous apelin injection in mothers increased the transplacental transport of glucose, and 4) intraperitoneal apelin administration in neonates increased glucose uptake in lung and muscle. Maternal food restriction drastically reduced apelinemia in both mothers and growth-restricted fetuses and altered the expression of the apelinergic system at the fetoplacental interface. Together, our data demonstrate that apelin controls fetal and neonatal glucose homeostasis and is altered by fetal growth restriction induced by maternal undernutrition.


Asunto(s)
Glucemia/metabolismo , Retardo del Crecimiento Fetal/genética , Feto/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Desnutrición/metabolismo , Complicaciones del Embarazo/metabolismo , Animales , Animales Recién Nacidos , Apelina , Receptores de Apelina , Glucemia/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Retardo del Crecimiento Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 3/genética , Homeostasis/efectos de los fármacos , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Placenta/metabolismo , Embarazo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Metabolism ; 62(3): 442-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23116519

RESUMEN

OBJECTIVE: Preeclampsia is a frequent and potentially lethal placental insufficiency pathology causing maternal hypertension and proteinuria, as well as a high rate of intrauterine growth retardation (IUGR) in offspring. Reduced nitric oxide (NO) production may play a role in the mechanisms of this disease. As exposure to adverse early life environment and IUGR has been proposed to increase cardiometabolic diseases risk, we investigated in rats the effects of maternal NO blockade on growth and metabolic phenotype of offspring. MATERIAL AND METHODS: Osmotic pumps were implanted in pregnant rats at E17 and diffused saline or L-NAME (50mg/day), a nitric oxide synthesis inhibitor. At birth, IUGR male newborns without limb defects were selected. Body growth, feeding behavior and glucose tolerance were evaluated in offspring. Organs weights, plasma level of several metabolic hormones and genes expressions were determined in fasted 9month-old rats. RESULTS: L-NAME mothers had elevated blood pressure at E20. Male offspring from L-NAME mothers had a markedly reduced birth weight and developed postnatal catch-up growth during lactation. Some L-NAME newborns presented some limb defects but were not selected in this study (1/3 of all pups). Improved glucose tolerance and hyperphagia after fasting were found in 3-month-old L-NAME rat but not thereafter. In 9-month-old L-NAME rats, a moderate increase of food intake during the light phase and, after fasting, an augmentation of plasma insulin and a reduction of brown adipose tissue (BAT) deposit were found associated with an increased expression of UCP-1 mRNA in this tissue. CONCLUSIONS: Despite IUGR and postnatal catch up growth, male rats exposed to L-NAME did not develop metabolic diseases when limb defects were not induced by L-NAME. We postulate that maternal hypertension during late gestation is not a major 'programming' metabolic factor for offspring.


Asunto(s)
Animales Recién Nacidos/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Hipertensión/metabolismo , Enfermedades Metabólicas/metabolismo , Complicaciones Cardiovasculares del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Prueba de Tolerancia a la Glucosa , Hipertensión/inducido químicamente , Masculino , Enfermedades Metabólicas/inducido químicamente , NG-Nitroarginina Metil Éster/administración & dosificación , Tamaño de los Órganos/fisiología , Embarazo , Complicaciones Cardiovasculares del Embarazo/inducido químicamente , Ratas , Ratas Wistar
15.
Peptides ; 35(1): 136-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22446510

RESUMEN

Apelin and its receptor APJ are expressed in fetal tissues but their function and regulation remain largely unknown. In rat, maternal treatment with a nitric oxide synthase inhibitor inducing hypertension was used to investigate apelin plasma levels in mother/fetus pairs and on the gene expression level of the apelin/APJ system in fetal tissues and placenta. At term, plasma levels of apelin were not modulated but APJ expression was increased in placenta and lung but reduced in heart. Apelin expression was increased only in the heart. We postulate that the apelinergic system may control fetal growth and cardiovascular functions in utero.


Asunto(s)
Hipertensión Inducida en el Embarazo/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Placenta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Apelina , Receptores de Apelina , Femenino , Corazón Fetal/metabolismo , Feto/metabolismo , Flujo Génico , Hipertensión Inducida en el Embarazo/inducido químicamente , Hipertensión Inducida en el Embarazo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Pulmón/metabolismo , NG-Nitroarginina Metil Éster , Especificidad de Órganos , Embarazo , Ratas , Receptores Acoplados a Proteínas G/genética
16.
Eur J Pharmacol ; 689(1-3): 278-84, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22683867

RESUMEN

Numerous data indicate that Rho kinase inhibitors, such as Fasudil, may constitute a novel therapy for cardiovascular and metabolic diseases. We evaluated long-term effects of exposure to Fasudil during late gestation (10 mg/day) in male rat offspring from birth until 9 months. We also analyzed its effects in offspring from hypertensive mothers treated with a nitric oxide synthesis inhibitor (L-NAME; 50 mg/day). Prenatal exposure to Fasudil did not affect birth weight, but increased body weight from postnatal day 7 (P7) to 9 months. In intrauterine growth-restricted (IUGR) fetuses exposed to L-NAME, maternal Fasudil treatment increased birth weight. At P42 and P180, rats exposed to Fasudil and L-NAME showed alterations of their food intake as well as an increased basal glycemia associated with mild glucose intolerance at 6 months which was also observed in Fasudil-exposed rats. In 9 month-old rats, exposure to Fasudil increased the daily food intake as well as hypothalamic mRNA level of the orexigenic NPY peptide without modulation of the anorexigenic POMC gene expression. Altogether, our data suggest that prenatal Fasudil exposure alleviates fetal growth in IUGR rats, but programs long-term metabolic disturbances including transient perturbations of glucose metabolism, a persistent increase of body weight gain, hyperphagia and an augmented expression of hypothalamic NPY orexigenic gene. We postulate that Fasudil treatment during perinatal periods may predispose individuals to the development of metabolic disorders.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Desarrollo Fetal/efectos de los fármacos , Hiperfagia/inducido químicamente , Sobrepeso/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/toxicidad , Factores de Edad , Animales , Femenino , Desarrollo Fetal/fisiología , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/prevención & control , Hiperfagia/metabolismo , Hiperfagia/fisiopatología , Masculino , Sobrepeso/metabolismo , Sobrepeso/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Distribución Aleatoria , Ratas , Ratas Wistar , Factores Sexuales
17.
PLoS One ; 7(11): e47986, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144842

RESUMEN

Males and females responses to gestational overnutrition set the stage for subsequent sex-specific differences in adult onset non communicable diseases. Placenta, as a widely recognized programming agent, contibutes to the underlying processes. According to our previous findings, a high-fat diet during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes. We further investigated the impact of diet and sex on placental histology, transcriptomic and epigenetic signatures in mice. Both basal gene expression and response to maternal high-fat diet were sexually dimorphic in whole placentas. Numerous genes showed sexually dimorphic expression, but only 11 genes regardless of the diet. In line with the key role of genes belonging to the sex chromosomes, 3 of these genes were Y-specific and 3 were X-specific. Amongst all the genes that were differentially expressed under a high-fat diet, only 16 genes were consistently affected in both males and females. The differences were not only quantitative but remarkably qualitative. The biological functions and networks of genes dysregulated differed markedly between the sexes. Seven genes of the epigenetic machinery were dysregulated, due to effects of diet, sex or both, including the Y- and X-linked histone demethylase paralogues Kdm5c and Kdm5d, which could mark differently male and female epigenomes. The DNA methyltransferase cofactor Dnmt3l gene expression was affected, reminiscent of our previous observation of changes in global DNA methylation. Overall, this striking sexual dimorphism of programming trajectories impose a considerable revision of the current dietary interventions protocols.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Epigénesis Genética , Expresión Génica , Placenta/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Histona Demetilasas , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Placenta/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Caracteres Sexuales , Transcriptoma
18.
Med Hypotheses ; 76(5): 726-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21377276

RESUMEN

Numerous data highlight the importance of a fine regulation of the fetal growth for the individual's subsequent susceptibility to diseases. Recent evidence suggests that neurotrophins may have important functions during pregnancy and that they could modulate fetal growth. We hypothesize that neurotrophins may participate in fetal growth by different pathways: (1) these molecules may participate in the development and the maturation of both central and peripheral fetal organs, including the placenta; (2) neurotrophins may constitute a link between the maternal nutrition and the fetal nutrients demand and thus could act as a nutrient sensor for the development and efficiency of the placenta; (3) maternal and placental neurotrophins may control the fetal development directly but also by activating the production and release of others growth factors. In conclusion, neurotrophins may participate to a new pathway controlling fetal growth and may be implicated, at least in part, under physiopathological conditions in disturbances of the fetal growth trajectory.


Asunto(s)
Desarrollo Fetal/fisiología , Factores de Crecimiento Nervioso/metabolismo , Placentación/fisiología , Líquido Amniótico/metabolismo , Animales , Femenino , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Modelos Biológicos , Modelos Teóricos , Placenta/fisiología , Embarazo
19.
Eur J Pharmacol ; 667(1-3): 402-9, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21514291

RESUMEN

Epidemiological and experimental data indicate that maternal undernutrition may sensitize the offspring to the apparition of chronic diseases such as metabolic syndrome and schizophrenia, suggesting that these pathologies may have a developmental origin. To test this hypothesis, we have compared the effects of a 4 weeks treatment of clozapine (30 mg/kg once daily, p.o.) or aripiprazole (10 mg/kg once daily, p.o.) on metabolic and hormonal parameters in 4-month-old male animals from control or 70% prenatally food-restricted mothers (FR30 model). Both neuroleptics did not markedly modify body weight gain and food intake in both controls and FR30 rats. Clozapine decreased insulin secretion in both groups but significantly diminished leptin, corticosterone and glucose plasma levels only in FR30 animals. Aripiprazole decreased corticosterone plasma levels only in FR30 animals. Using quantitative RT-PCR array containing 84 obesity-related genes, we identified several genes involved in energy metabolism regulation whose expression was modified by clozapine or aripiprazole in adult male rat hypothalami. In addition, we demonstrated that expression of some of these genes was differentially affected by each neuroleptic in the hypothalamus of both FR30 and control animals. Although no marked metabolic alterations were observed in both control and FR30 animals after clozapine or aripiprazole treatment, our data indicate that offspring from undernourished mothers exhibit a modified sensitivity to atypical neuroleptics. Our results do not rule out a putative developmental origin of schizophrenia and may help to understand the way by which atypical neuroleptics, such as clozapine, sensitize schizophrenic patients to the development of metabolic disorders.


Asunto(s)
Antipsicóticos/farmacología , Clozapina/farmacología , Desnutrición , Sistema Nervioso/crecimiento & desarrollo , Piperazinas/farmacología , Fenómenos Fisiologicos de la Nutrición Prenatal , Quinolonas/farmacología , Esquizofrenia/patología , Animales , Aripiprazol , Peso Corporal/efectos de los fármacos , Susceptibilidad a Enfermedades/inducido químicamente , Ingestión de Alimentos/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormonas/sangre , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Sistema Nervioso/efectos de los fármacos , Obesidad/genética , Embarazo , Ratas , Ratas Wistar , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología
20.
PLoS One ; 5(12): e14398, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21200436

RESUMEN

BACKGROUND: Changes in imprinted gene dosage in the placenta may compromise the prenatal control of nutritional resources. Indeed monoallelic behaviour and sensitivity to changes in regional epigenetic state render imprinted genes both vulnerable and adaptable. METHODS AND FINDINGS: We investigated whether a high-fat diet (HFD) during pregnancy modified the expression of imprinted genes and local and global DNA methylation patterns in the placenta. Pregnant mice were fed a HFD or a control diet (CD) during the first 15 days of gestation. We compared gene expression patterns in total placenta homogenates, for male and female offspring, by the RT-qPCR analysis of 20 imprinted genes. Sexual dimorphism and sensitivity to diet were observed for nine genes from four clusters on chromosomes 6, 7, 12 and 17. As assessed by in situ hybridization, these changes were not due to variation in the proportions of the placental layers. Bisulphite-sequencing analysis of 30 CpGs within the differentially methylated region (DMR) of the chromosome 17 cluster revealed sex- and diet-specific differential methylation of individual CpGs in two conspicuous subregions. Bioinformatic analysis suggested that these differentially methylated CpGs might lie within recognition elements or binding sites for transcription factors or factors involved in chromatin remodelling. Placental global DNA methylation, as assessed by the LUMA technique, was also sexually dimorphic on the CD, with lower methylation levels in male than in female placentae. The HFD led to global DNA hypomethylation only in female placenta. Bisulphite pyrosequencing showed that neither B1 nor LINE repetitive elements could account for these differences in DNA methylation. CONCLUSIONS: A HFD during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes important in the control of many cellular, metabolic and physiological functions potentially involved in adaptation and/or evolution. These findings highlight the importance of studying both sexes in epidemiological protocols and dietary interventions.


Asunto(s)
Alimentación Animal , Metilación de ADN , Grasas de la Dieta/metabolismo , Regulación de la Expresión Génica , Impresión Genómica , Placenta/metabolismo , Alelos , Animales , Epigénesis Genética , Femenino , Dosificación de Gen , Masculino , Ratones , Embarazo , Preñez , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA