Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Kidney J ; 13(3): 371-379, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32699617

RESUMEN

BACKGROUND: Posterior urethral valves (PUV) account for 17% of paediatric end-stage renal disease. A major issue in the management of PUV is prenatal prediction of postnatal renal function. Fetal ultrasound and fetal urine biochemistry are currently employed for this prediction, but clearly lack precision. We previously developed a fetal urine peptide signature that predicted in utero with high precision postnatal renal function in fetuses with PUV. We describe here the objectives and design of the prospective international multicentre ANTENATAL (multicentre validation of a fetal urine peptidome-based classifier to predict postnatal renal function in posterior urethral valves) study, set up to validate this fetal urine peptide signature. METHODS: Participants will be PUV pregnancies enrolled from 2017 to 2021 and followed up until 2023 in >30 European centres endorsed and supported by European reference networks for rare urological disorders (ERN eUROGEN) and rare kidney diseases (ERN ERKNet). The endpoint will be renal/patient survival at 2 years postnatally. Assuming α = 0.05, 1-ß = 0.8 and a mean prevalence of severe renal outcome in PUV individuals of 0.35, 400 patients need to be enrolled to validate the previously reported sensitivity and specificity of the peptide signature. RESULTS: In this largest multicentre study of antenatally detected PUV, we anticipate bringing a novel tool to the clinic. Based on urinary peptides and potentially amended in the future with additional omics traits, this tool will be able to precisely quantify postnatal renal survival in PUV pregnancies. The main limitation of the employed approach is the need for specialized equipment. CONCLUSIONS: Accurate risk assessment in the prenatal period should strongly improve the management of fetuses with PUV.

2.
J Bone Miner Res ; 30(8): 1369-76, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25656438

RESUMEN

Marfan syndrome (MFS) is a rare connective tissue disorder caused by mutation in the gene encoding the extracellular matrix protein fibrillin-1 (FBN1), leading to transforming growth factor-beta (TGF-ß) signaling dysregulation. Although decreased axial and peripheral bone mineral density (BMD) has been reported in adults with MFS, data about the evolution of bone mass during childhood and adolescence are limited. The aim of the present study was to evaluate bone and muscle characteristics in children, adolescents, and young adults with MFS. The study population included 48 children and young adults (22 girls) with MFS with a median age of 11.9 years (range 5.3 to 25.2 years). The axial skeleton was analyzed at the lumbar spine using dual-energy X-ray absorptiometry (DXA), whereas the appendicular skeleton (hand) was evaluated using the BoneXpert system (with the calculation of the Bone Health Index). Muscle mass was measured by DXA. Compared with healthy age-matched controls, bone mass at the axial and appendicular levels and muscle mass were decreased in children with MFS and worsened from childhood to adulthood. Vitamin D deficiency (<50 nmol/L) was found in about a quarter of patients. Serum vitamin D levels were negatively correlated with age and positively correlated with lumbar spine areal and volumetric BMD. Lean body mass (LBM) Z-scores were positively associated with total body bone mineral content (TB-BMC) Z-scores, and LBM was an independent predictor of TB-BMC values, suggesting that muscle hypoplasia could explain at least in part the bone loss in MFS. Patients with a FBN1 premature termination codon mutation had a more severe musculoskeletal phenotype than patients with an inframe mutation, suggesting the involvement of TGF-ß signaling dysregulation in the pathophysiologic mechanisms. In light of these results, we recommend that measurement of bone mineral status should be part of the longitudinal clinical investigation of MFS children.


Asunto(s)
Envejecimiento , Genotipo , Síndrome de Marfan , Proteínas de Microfilamentos/genética , Músculo Esquelético , Columna Vertebral , Absorciometría de Fotón , Adulto , Envejecimiento/genética , Envejecimiento/patología , Densidad Ósea , Niño , Preescolar , Codón de Terminación , Estudios Transversales , Femenino , Fibrilina-1 , Fibrilinas , Estudios de Seguimiento , Humanos , Masculino , Síndrome de Marfan/diagnóstico por imagen , Síndrome de Marfan/genética , Síndrome de Marfan/fisiopatología , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/fisiopatología , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/fisiopatología
3.
Eur J Hum Genet ; 23(2): 252-5, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24916642

RESUMEN

The SNORD116 locus lies in the 15q11-13 region of paternally expressed genes implicated in Prader-Willi Syndrome (PWS), a complex disease accompanied by obesity and severe neurobehavioural disturbances. Cases of PWS patients with a deletion encompassing the SNORD116 gene cluster, but preserving the expression of flanking genes, have been described. We report a 23-year-old woman who presented clinical criteria of PWS, including the behavioural and nutritional features, obesity, developmental delay and endocrine dysfunctions with hyperghrelinemia. We found a paternally transmitted highly restricted deletion of the SNORD116 gene cluster, the shortest described to date (118 kb). This deletion was also present in the father. This finding in a human case strongly supports the current hypothesis that lack of the paternal SNORD116 gene cluster has a determinant role in the pathogenesis of PWS. Moreover, targeted analysis of the SNORD116 gene cluster, complementary to SNRPN methylation analysis, should be carried out in subjects with a phenotype suggestive of PWS.


Asunto(s)
Eliminación de Gen , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , Adulto , Femenino , Humanos , Masculino , Síndrome de Prader-Willi/diagnóstico , ARN Nucleolar Pequeño/metabolismo
4.
Biochim Biophys Acta ; 1831(1): 93-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22867754

RESUMEN

LPA and LPA(1) have been shown to increase osteoblastic proliferation and differentiation as well as activation of osteoclasts. Cell and animal model studies have suggested that LPA is produced by bone cells and bone tissues. We obtained data from invalidated mice which support the hypothesis that LPA(1) is involved in bone development by promoting osteogenesis. LPA(1)-invalidated mice demonstrate growth and sternal and costal abnormalities, which highlights the specific roles of LPA(1) during bone development. Microcomputed tomography and histological analysis demonstrate osteoporosis in the trabecular and cortical bone of LPA(1)-invalidated mice. Moreover, bone marrow mesenchymal progenitors from these mice displayed decreased osteoblastic differentiation. Infrared analysis did not indicate osteomalacia in the bone tissue of LPA(1)-invalidated mice. LPA(1) displays opposite effects to LPA(4) on the related G proteins G(i) and G(s), responsible for decrease and increase of the cAMP level respectively, which itself is essential to the control of osteoblastic differentiation. The opposite effects of LPA(1) and LPA(4) during osteoblastic differentiation support the possibility that new pharmacological agents derived from the LPA pathways could be found and used in clinical practice to positively influence bone formation and treat osteoporosis. The paracrine effect of LPA is potentially modulated by its concentration in bone tissues, which may result from various intracellular and extracellular pathways. The relevance of LPA(1) in bone remodeling, as a receptor able to influence both osteoblast and osteoclast activity, still deserves further clarification. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Asunto(s)
Huesos/metabolismo , Huesos/patología , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Huesos/diagnóstico por imagen , Lisofosfolípidos/biosíntesis , Ratones , Ratones Transgénicos , Modelos Biológicos , Osteoblastos/metabolismo , Osteoblastos/patología , Fenotipo , Radiografía , Transducción de Señal
5.
Bone ; 49(3): 395-403, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21569876

RESUMEN

Lysophosphatidic acid (LPA) is a lipid mediator that acts in paracrine systems via interaction with a subset of G protein-coupled receptors (GPCRs). LPA promotes cell growth and differentiation, and has been shown to be implicated in a variety of developmental and pathophysiological processes. At least 6 LPA GPCRs have been identified to date: LPA1-LPA6. Several studies have suggested that local production of LPA by tissues and cells contributes to paracrine regulation, and a complex interplay between LPA and its receptors, LPA1 and LPA4, is believed to be involved in the regulation of bone cell activity. In particular, LPA1 may activate both osteoblasts and osteoclasts. However, its role has not as yet been examined with regard to the overall status of bone in vivo. We attempted to clarify this role by defining the bone phenotype of LPA1((-/-)) mice. These mice demonstrated significant bone defects and low bone mass, indicating that LPA1 plays an important role in osteogenesis. The LPA1((-/-)) mice also presented growth and sternal and costal abnormalities, which highlights the specific roles of LPA1 during bone development. Microcomputed tomography and histological analysis demonstrated osteoporosis in the trabecular and cortical bone of LPA1((-/-)) mice. Finally, bone marrow mesenchymal progenitors from these mice displayed decreased osteoblastic differentiation. These results suggest that LPA1 strongly influences bone development both qualitatively and quantitatively and that, in vivo, its absence results in decreased osteogenesis with no clear modification of osteoclasis. They open perspectives for a better understanding of the role of the LPA/LPA1 paracrine pathway in bone pathophysiology.


Asunto(s)
Huesos/anomalías , Huesos/fisiología , Diferenciación Celular/fisiología , Osteogénesis/fisiología , Isoformas de Proteínas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Biomarcadores/metabolismo , Densidad Ósea , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Huesos/anatomía & histología , Huesos/diagnóstico por imagen , Lisofosfolípidos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estado Nutricional , Osteoblastos/citología , Osteoblastos/fisiología , Osteoclastos/citología , Osteoclastos/fisiología , Isoformas de Proteínas/genética , Receptores del Ácido Lisofosfatídico/genética , Microtomografía por Rayos X
6.
Mol Cell Biol ; 30(10): 2498-507, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20308328

RESUMEN

LEOPARD syndrome (LS), a disorder with multiple developmental abnormalities, is mainly due to mutations that impair the activity of the tyrosine phosphatase SHP2 (PTPN11). How these alterations cause the disease remains unknown. We report here that fibroblasts isolated from LS patients displayed stronger epidermal growth factor (EGF)-induced phosphorylation of both AKT and glycogen synthase kinase 3beta (GSK-3beta) than fibroblasts from control patients. Similar results were obtained in HEK293 cells expressing LS mutants of SHP2. We found that the GAB1/phosphoinositide 3-kinase (PI3K) complex was more abundant in fibroblasts from LS than control subjects and that both AKT and GSK-3beta hyperphosphorylation were prevented by reducing GAB1 expression or by overexpressing a GAB1 mutant unable to bind to PI3K. Consistently, purified recombinant LS mutants failed to dephosphorylate GAB1 PI3K-binding sites. These mutants induced PI3K-dependent increase in cell size in a model of chicken embryo cardiac explants and in transcriptional activity of the atrial natriuretic factor (ANF) gene in neonate rat cardiomyocytes. In conclusion, SHP2 mutations causing LS facilitate EGF-induced PI3K/AKT/GSK-3beta stimulation through impaired GAB1 dephosphorylation, resulting in deregulation of a novel signaling pathway that could be involved in LS pathology.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Síndrome LEOPARD , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Células Cultivadas , Embrión de Pollo , Activación Enzimática , Fibroblastos/citología , Fibroblastos/fisiología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Síndrome LEOPARD/genética , Síndrome LEOPARD/metabolismo , Síndrome LEOPARD/patología , Mutación , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
J Clin Invest ; 119(12): 3830-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19907080

RESUMEN

Growth hormone (GH) is a major metabolic regulator that functions by stimulating lipolysis, preventing protein catabolism, and decreasing insulin-dependent glucose disposal. Modulation of hepatic sensitivity to GH and the downstream effects on the GH/IGF1 axis are important events in the regulation of metabolism in response to variations in food availability. For example, during periods of reduced nutrient availability, the liver becomes resistant to GH actions. However, the mechanisms controlling hepatic GH resistance are currently unknown. Here, we investigated the role of 2 tetraspanning membrane proteins, leptin receptor overlapping transcript (LEPROT; also known as OB-RGRP) and LEPROT-like 1 (LEPROTL1), in controlling GH sensitivity. Transgenic mice expressing either human LEPROT or human LEPROTL1 displayed growth retardation, reduced plasma IGF1 levels, and impaired hepatic sensitivity to GH, as measured by STAT5 phosphorylation and Socs2 mRNA expression. These phenotypes were accentuated in transgenic mice expressing both proteins. Moreover, gene silencing of either endogenous Leprot or Leprotl1 in H4IIE hepatocytes increased GH signaling and enhanced cell-surface GH receptor. Importantly, we found that both LEPROT and LEPROTL1 expression were regulated in the mouse liver by physiologic and pathologic changes in glucose homeostasis. Together, these data provide evidence that LEPROT and LEPROTL1 influence liver GH signaling and that regulation of the genes encoding these proteins may constitute a molecular link between nutritional signals and GH actions on body growth and metabolism.


Asunto(s)
Proteínas Portadoras/metabolismo , Hormona del Crecimiento/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Ayuno/metabolismo , Femenino , Hormona del Crecimiento/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Somatotropina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética
8.
J Clin Endocrinol Metab ; 93(7): 2800-5, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18460565

RESUMEN

BACKGROUND: High plasma ghrelin levels have been reported in Prader-Willi syndrome (PWS). However, little is known about plasma ghrelin in these children during the first years of life characterized by a failure to thrive. OBJECTIVE: The objective of the study was to investigate total plasma ghrelin levels in children with PWS and controls from 2 months to 17 years. SUBJECTS AND METHODS: Forty children with PWS [24 boys, 16 girls, median age 3.6 yr, median body mass index (BMI) Z-score 0.3] were compared with 84 controls (57 boys, 27 girls, median age 4.2 yr median BMI Z-score 0.1). Children were then divided into two groups according to age and GH treatment. RESULTS: Median plasma ghrelin levels were significantly higher in children with PWS, compared with controls at any age (568 vs. 173, P < 0.0001) and decreased with age in both groups (P < 0.0001). In the whole group of PWS, we found an inverse relationship between ghrelin and BMI Z-score, insulin, homeostasis model assessment insulin resistance index, leptin, and lean mass. Plasma ghrelin levels were higher in children with PWS than controls, both in the youngest children below 3 yr who were not receiving GH (771 vs. 233, P < 0.0001) and in the children older than 3 yr, all of whom were treated with GH (428 vs. 159, P < 0.0001). CONCLUSIONS: Plasma ghrelin levels in children with PWS are elevated at any age, including during the first years of life, thus preceding the development of obesity.


Asunto(s)
Ghrelina/sangre , Obesidad/sangre , Síndrome de Prader-Willi/sangre , Adolescente , Factores de Edad , Niño , Preescolar , Femenino , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Lactante , Masculino , Síndrome de Prader-Willi/tratamiento farmacológico
9.
Cell Tissue Res ; 329(2): 283-94, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17443352

RESUMEN

The dental follicle (DF) surrounding the developing tooth germ is an ectomesenchymal tissue composed of various cell populations derived from the cranial neural crest. Human dental follicle cells (HDFC) are believed to contain precursor cells for cementoblasts, periodontal ligament cells, and osteoblasts. Bone morphogenetic proteins (BMPs) produced by Hertwig's epithelial root sheath or present in enamel matrix derivatives (EMD) seem to be involved in the control of DF cell differentiation, but their precise function remains largely unknown. We report the immunolocalization of STRO-1 (a marker of multipotential mesenchymal progenitor cells) and BMP receptors (BMPR) in DF in vivo. In culture, HDFC co-express STRO-1/BMPR and exhibit multilineage properties. Incubation with rhBMP-2 and rhBMP-7 or EMD for 24 h increases the expression of BMP-2 and BMP-7 by HDFC. Long-term stimulation of these cells by rhBMP-2 and/or rhBMP-7 or EMD significantly increases alkaline phosphatase activity (AP) and mineralization. Expression of cementum attachment protein (CAP) and cementum protein-23 (CP-23), two putative cementoblast markers, has been detected in EMD-stimulated whole DF and in cultured HDFC stimulated with EMD or BMP-2 and BMP-7. RhNoggin, a BMP antagonist, abolishes AP activity, mineralization, and CAP/CP-23 expression in HDFC cultures and the expression of BMP-2 and BMP-7 induced by EMD. Phosphorylation of Smad-1 and MAPK is stimulated by EMD or rhBMP-2. However, rhNoggin blocks only Smad-1 phosphorylation under these conditions. Thus, EMD may activate HDFC toward the cementoblastic phenotype, an effect mainly (but not exclusively) involving both exogenous and endogenous BMP-dependent pathways.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Cemento Dental/fisiología , Proteínas del Esmalte Dental/fisiología , Saco Dental/fisiología , Células Madre Mesenquimatosas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Adolescente , Fosfatasa Alcalina/biosíntesis , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 7 , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/farmacología , Calcificación Fisiológica , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Niño , Cemento Dental/metabolismo , Proteínas del Esmalte Dental/biosíntesis , Proteínas del Esmalte Dental/farmacología , Saco Dental/citología , Saco Dental/metabolismo , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosforilación , Proteínas Recombinantes/farmacología , Proteína Smad1/metabolismo , Proteína Smad1/fisiología , Técnicas de Cultivo de Tejidos , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/farmacología
10.
Horm Res ; 62(1): 49-54, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15192277

RESUMEN

BACKGROUND: Elevated plasma ghrelin levels have recently been reported in adults and children with Prader-Willi syndrome (PWS). The aim of the study is to investigate the relationship between obesity, growth hormone (GH) deficiency (GHD) and ghrelinemia in PWS and to examine whether hyperghrelinemia is specific to PWS. METHODS: We measured fasting ghrelinemia in children with PWS, idiopathic GHD (iGHD), obese children, controls and in 6 children presenting another congenital syndrome associated with GHD: pituitary stalk interruption (PSI). RESULTS: Children with PWS exhibited significantly higher ghrelin levels (995 pg/ml (801/1,099, median 1st/3rd quartile)) than iGHD (517 pg/ml (392/775)), obese (396 pg/ml (145/610)) and control (605 ng/ml (413/753)) children. Similar to PWS hyperghrelinemia was found in PSI children (1,029 pg/ml (705/1,151)), and was not modified by GH treatment. CONCLUSION: We conclude that hyperghrelinemia in PWS and PSI is not related to GH secretion. We hypothesize that a major site of ghrelin action is at the hypothalamic level and that a 'ghrelin resistance' syndrome may be present in these patients, primarily due to a hypothalamic defect. Combined alterations such as impaired serotonin receptor regulation associated with abnormal ghrelin responsiveness are probably responsible for obesity in PWS.


Asunto(s)
Modelos Biológicos , Hormonas Peptídicas/sangre , Hipófisis/anomalías , Síndrome de Prader-Willi/sangre , Envejecimiento/sangre , Índice de Masa Corporal , Estudios de Casos y Controles , Niño , Anomalías Congénitas/sangre , Femenino , Ghrelina , Hormona del Crecimiento/uso terapéutico , Hormona de Crecimiento Humana/deficiencia , Hormona de Crecimiento Humana/metabolismo , Humanos , Masculino , Obesidad/sangre , Síndrome de Prader-Willi/tratamiento farmacológico , Síndrome de Prader-Willi/metabolismo , Errores Congénitos del Metabolismo Esteroideo/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...