Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 25(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38256140

RESUMEN

Members of the transforming growth factor ß (TGF-ß) family have been implicated in the biology of several cancers. In this review, we focus on the role of TGFß and bone morphogenetic protein (BMP) signaling in glioblastoma. Glioblastoma (GBM) is the most common malignant brain tumor in adults; it presents at a median age of 64 years, but can occur at any age, including childhood. Unfortunately, there is no cure, and even patients undergoing current treatments (surgical resection, radiotherapy, and chemotherapy) have a median survival of 15 months. There is a great need to identify new therapeutic targets to improve the treatment of GBM patients. TGF-ßs signaling promotes tumorigenesis in glioblastoma, while BMPs suppress tumorigenic potential by inducing tumor cell differentiation. In this review, we discuss the actions of TGF-ßs and BMPs on cancer cells as well as in the tumor microenvironment, and their use in potential therapeutic intervention.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Proteínas de la Superfamilia TGF-beta , Humanos , Neoplasias Encefálicas/genética , Carcinogénesis , Diferenciación Celular , Glioblastoma/genética , Factor de Crecimiento Transformador beta , Microambiente Tumoral , Proteínas de la Superfamilia TGF-beta/genética
2.
Gene ; 787: 145627, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-33831497

RESUMEN

The objective of this study was to identify the gonadal somatic cells in the Yesso scallop using a novel molecular marker. This study is the first to identify the bone morphogenetic protein 2a (Bmp2a) gene as a gonadal somatic cell-specific gene in this bivalve. We performed a transcriptomic survey to identify the transforming growth factor-ß (TGFß) superfamily members that act in Yesso scallop gonad development. BLAST survey, phylogenetic tree, and RT-PCR analyses screened BMP molecules (i.e., bmp2a and bmp10a), which are members of the TGFß superfamily that show gonad-specific expression. Among the BMPs from the Yesso scallop, in situ hybridization accompanied by RNAscope assay identified that bmp2a mRNA was specifically expressed in the gonadal somatic cells localized in the interspace between germ cells. Real-time quantitative PCR (qPCR) analysis revealed that bmp2a mRNA expression increased during the reproductive phase. The relative expression of bmp2a mRNA was lowest at the beginning of the growing stage and peaked at the mature stage in both sexes. These observations indicate that bmp2a-positive gonadal somatic cells support germ cell growth and differentiation during the reproductive phase for both sexes. This study provides new insights into gonadal somatic cell biology in marine invertebrates and we propose that TGFß signaling is necessary for gonad development in bivalves.


Asunto(s)
Gónadas/citología , Gónadas/metabolismo , Pectinidae/metabolismo , Proteínas de la Superfamilia TGF-beta/metabolismo , Animales , Antígenos de Diferenciación , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Simulación por Computador , Femenino , Marcadores Genéticos , Gónadas/crecimiento & desarrollo , Hibridación in Situ , Masculino , Pectinidae/citología , Pectinidae/genética , Pectinidae/crecimiento & desarrollo , Filogenia , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducción , Transducción de Señal , Proteínas de la Superfamilia TGF-beta/genética , Distribución Tisular , Transcriptoma
3.
Genes (Basel) ; 11(3)2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32213808

RESUMEN

During embryonic development in vertebrates, morphogens play an important role in cell fate determination and morphogenesis. Bone morphogenetic proteins (BMPs) belonging to the transforming growth factor-ß (TGF-ß) family control the dorsal-ventral (DV) patterning of embryos, whereas other morphogens such as fibroblast growth factor (FGF), Wnt family members, and retinoic acid (RA) regulate the formation of the anterior-posterior (AP) axis. Activation of morphogen signaling results in changes in the expression of target genes including transcription factors that direct cell fate along the body axes. To ensure the correct establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated by a fine-tuning of morphogen signaling. In this review, we focus on the interplay of various intracellular regulatory mechanisms and discuss how communication among morphogen signaling pathways modulates body axis formation in vertebrate embryos.


Asunto(s)
Tipificación del Cuerpo , Comunicación Celular , Proteínas de la Superfamilia TGF-beta/metabolismo , Proteínas Wnt/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de la Superfamilia TGF-beta/genética , Proteínas Wnt/genética
4.
Domest Anim Endocrinol ; 70: 106398, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31677486

RESUMEN

The aim of this study was to investigate some of the growth and transcriptional factors originating from oocytes and granulosa cells in follicular fluid and to identify the relationships between the basic blood metabolite-metabolic hormones and intrafollicular lipopolysaccharide (LPS) concentrations. Thirty cows included in the study were allocated into 2 groups comprising 15 cows with healthy preovulatory follicles (cyclic cows) and 15 cows with confirmed cystic follicles. The ovaries and uteri of all cows were assessed by transrectal ultrasonographic examination. Blood serum samples were collected at 15, 25, 35, 45, and 55 d after calving for analysis of nonesterified fatty acids, ß-hydroxybutyrate, insulin, glucose, IGF-I, ACTH, and cortisol. Ovaries and uteri were examined using transrectal ultrasound. Vaginal discharge was evaluated on the same days. Follicular fluid was also aspirated on days 35-55 from the healthy preovulatory follicles and cystic follicles using a transvaginal ovum pickup method. The densitometric levels of inhibin-α, growth and differentiation factor (GDF-9), bone morphogenetic protein (BMP-6), and GATA-4 and GATA-6 proteins were analyzed by the Western blotting technique; the concentrations of antimullerian hormone (AMH), IGF-I, estradiol-17 beta (E2), and progesterone (P4) were determined by ELISA; and the concentrations of LPS in the follicular fluid were measured by the Limulus amebocyte lysate test. The serum insulin, ACTH, and cortisol concentrations were higher in cystic cows than cyclic cows, but serum IGF-I concentrations were lower in cystic cows. The IGF-I concentrations of cystic follicular fluids were lower, whereas AMH levels were significantly greater than those of healthy preovulatory follicular fluids. The cystic follicles had significantly lower expression levels of GDF-9, BMP-6, GATA-4, and GATA-6; in contrast, inhibin-α expression and LPS concentrations were significantly higher than in healthy preovulatory follicles. The proportion of pathologic vaginal discharge within 25 d postpartum in cystic cows were higher than in the cyclic group. In conclusion, it is suggested that intrafollicular dysregulation of the transforming growth factor-ß superfamily, growth, and transcriptional factors is affected by high intrafollicular LPS concentrations and systemic metabolic changes and these disturbances may be responsible for the generation of ovarian cysts.


Asunto(s)
Enfermedades de los Bovinos/metabolismo , Lipopolisacáridos/metabolismo , Quistes Ováricos/veterinaria , Folículo Ovárico/crecimiento & desarrollo , Proteínas de la Superfamilia TGF-beta/metabolismo , Factores de Transcripción/metabolismo , Animales , Glucemia , Bovinos , Enfermedades de los Bovinos/sangre , Femenino , Regulación de la Expresión Génica , Quistes Ováricos/metabolismo , Proteínas de la Superfamilia TGF-beta/genética , Factores de Transcripción/genética
5.
Genomics ; 112(1): 332-345, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30779940

RESUMEN

Guard hair and cashmere undercoat are developed from primary and secondary hair follicle, respectively. Little is known about the gene expression differences between primary and secondary hair follicle cycling. In this study, we obtained RNA-seq data from cashmere and milk goats grown at four different seasons. We studied the differentially expressed genes (DEGs) during the yearly hair follicle cycling, and between cashmere and milk goats. WNT, NOTCH, MAPK, BMP, TGFß and Hedgehog signaling pathways were involved in hair follicle cycling in both cashmere and milk goat. However, Milk goat DEGs between different months were significantly more than cashmere goat DEGs, with the largest difference being identified in December. Some expression dynamics were confirmed by quantitative PCR and western blot, and immunohistochemistry. This study offers new information sources related to hair follicle cycling in milk and cashmere goats, which could be applicable to improve the wool production and quality.


Asunto(s)
Cabras/genética , Folículo Piloso/metabolismo , Transcriptoma , Animales , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Genómica , Cabras/metabolismo , Folículo Piloso/crecimiento & desarrollo , RNA-Seq , Estaciones del Año , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 3022-3037, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29928977

RESUMEN

The pathomechanisms underlying oxidative phosphorylation (OXPHOS) diseases are not well-understood, but they involve maladaptive changes in mitochondria-nucleus communication. Many studies on the mitochondria-nucleus cross-talk triggered by mitochondrial dysfunction have focused on the role played by regulatory proteins, while the participation of miRNAs remains poorly explored. MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) is mostly caused by mutation m.3243A>G in mitochondrial tRNALeu(UUR) gene. Adverse cardiac and neurological events are the commonest causes of early death in m.3243A>G patients. Notably, the incidence of major clinical features associated with this mutation has been correlated to the level of m.3243A>G mutant mitochondrial DNA (heteroplasmy) in skeletal muscle. In this work, we used a transmitochondrial cybrid model of MELAS (100% m.3243A>G mutant mitochondrial DNA) to investigate the participation of miRNAs in the mitochondria-nucleus cross-talk associated with OXPHOS dysfunction. High-throughput analysis of small-RNA-Seq data indicated that expression of 246 miRNAs was significantly altered in MELAS cybrids. Validation of selected miRNAs, including miR-4775 and miR-218-5p, in patient muscle samples revealed miRNAs whose expression declined with high levels of mutant heteroplasmy. We show that miR-218-5p and miR-4775 are direct regulators of fetal cardiac genes such as NODAL, RHOA, ISL1 and RXRB, which are up-regulated in MELAS cybrids and in patient muscle samples with heteroplasmy above 60%. Our data clearly indicate that TGF-ß superfamily signaling and an epithelial-mesenchymal transition-like program are activated in MELAS cybrids, and suggest that down-regulation of miRNAs regulating fetal cardiac genes is a risk marker of heart failure in patients with OXPHOS diseases.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Insuficiencia Cardíaca/genética , Síndrome MELAS/genética , MicroARNs/genética , Miocardio/patología , ARN de Transferencia de Leucina/genética , Línea Celular Tumoral , ADN Mitocondrial/genética , Conjuntos de Datos como Asunto , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Insuficiencia Cardíaca/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome MELAS/complicaciones , Síndrome MELAS/patología , MicroARNs/metabolismo , Mitocondrias/genética , Mitocondrias/patología , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Miocardio/citología , Miocardio/metabolismo , Fosforilación Oxidativa , Análisis de Secuencia de ARN , Transducción de Señal/genética , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Regulación hacia Arriba
7.
J Heart Valve Dis ; 25(6): 708-715, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-28290170

RESUMEN

BACKGROUND AND AIM OF THE STUDY: Pulmonary arterial hypertension (PAH) is a common accompaniment of rheumatic mitral stenosis (MS), with 70% of patients showing evidence of different grades of PAH. The latter condition is found to be a prognostic factor influencing disease outcome even after interventional or surgical therapy. The cause of the non-regression of PAH following successful balloon mitral valvotomy (BMV) is not clear. Hence, the study aim was to determine if there is an association of mutations in the genes of the TGF-ß superfamily and non-regression of PAH in patients who undergo a successful BMV. METHODS: Forty-six patients who underwent BMV and fulfilled the recruitment criteria were enrolled prospectively in this case-control study. Among the patients, 27 had non-regression of PAH while 19 had regression of PAH and served as controls. The mean age of the population was 32.63 ± 10.65 years. RESULTS: No statistically significant differences were identified in any of the baseline parameters between the two groups. None of the samples had BMPR2 or ACVRL1 mutations. Ten of the patients and four of the controls were positive for Endoglin mutation, but the inter-group difference was not statistically significant (p = 0.25) CONCLUSIONS: The present study - the first of its kind - showed that deletion-duplication mutations in the BMPR2 or ACVRL1 genes may not be associated with non-regression of PAH, even after successful BMV, or in a wider sense serve as a contributor to PAH in rheumatic MS. The association of Endoglin mutation and non-regression of PAH warrants further investigation in a larger population.


Asunto(s)
Valvuloplastia con Balón , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Estenosis de la Válvula Mitral/cirugía , Cardiopatía Reumática/cirugía , Proteínas de la Superfamilia TGF-beta/genética , Receptores de Activinas Tipo II/genética , Adulto , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Estudios de Casos y Controles , Endoglina/genética , Femenino , Eliminación de Gen , Reordenamiento Génico , Humanos , Masculino , Estenosis de la Válvula Mitral/fisiopatología , Proyectos Piloto , Estudios Prospectivos , Cardiopatía Reumática/fisiopatología , Adulto Joven
8.
PLoS One ; 10(12): e0145322, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26683658

RESUMEN

Genomic technologies including microarrays and next-generation sequencing have enabled the generation of molecular signatures of prostate cancer. Lists of differentially expressed genes between malignant and non-malignant states are thought to be fertile sources of putative prostate cancer biomarkers. However such lists of differentially expressed genes can be highly variable for multiple reasons. As such, looking at differential expression in the context of gene sets and pathways has been more robust. Using next-generation genome sequencing data from The Cancer Genome Atlas, differential gene expression between age- and stage- matched human prostate tumors and non-malignant samples was assessed and used to craft a pathway signature of prostate cancer. Up- and down-regulated genes were assigned to pathways composed of curated groups of related genes from multiple databases. The significance of these pathways was then evaluated according to the number of differentially expressed genes found in the pathway and their position within the pathway using Gene Set Enrichment Analysis and Signaling Pathway Impact Analysis. The "transforming growth factor-beta signaling" and "Ran regulation of mitotic spindle formation" pathways were strongly associated with prostate cancer. Several other significant pathways confirm reported findings from microarray data that suggest actin cytoskeleton regulation, cell cycle, mitogen-activated protein kinase signaling, and calcium signaling are also altered in prostate cancer. Thus we have demonstrated feasibility of pathway analysis and identified an underexplored area (Ran) for investigation in prostate cancer pathogenesis.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Anciano , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/genética , Transducción de Señal , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Transcriptoma
9.
Mol Ecol ; 24(18): 4664-78, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26268413

RESUMEN

The Antarctic icefish, a family (Channichthyidae) of teleosts within the perciform suborder Notothenioidei, are the only known vertebrates without oxygen-transporting haemoglobins and that are largely devoid of circulating erythrocytes. To elucidate the evo-devo mechanisms underpinning the suppressed erythropoiesis in the icefish, we conducted comparative studies on the transcriptomes and microRNAomes of the primary haematopoietic tissues between an icefish (Chionodraco hamatus) and two red-blooded notothenioids (Trematomus bernacchii and Gymnodraco acuticeps). We identified substantial remodelling of the haematopoietic programs in the icefish through which erythropoiesis is selectively suppressed. Experimental verification showed that erythropoietic suppression in the icefish may be attributable to the upregulation of TGF-ß signalling, which coincides with reductions in multiple transcription factors essential for erythropoiesis and the upregulation of hundreds of microRNAs, the majority (> 80%) of which potentially target erythropoiesis regulating factors. Of the six microRNAs selected for verification, three miRNAs (miR-152, miR-1388 and miR-16b) demonstrated suppressive functions on GATA1 and ALAS2, which are two factors important for erythroid differentiation, resulting in reduced numbers of erythroids in microinjected zebra fish embryos. Codon substitution analyses of the genes of the TGF-ß superfamily revealed signs of positive selection in TGF-ß1 and endoglin in the lineages leading to Antarctic notothenioids. Both genes are previously known to function in erythropoietic suppression. These findings implied a general trend of erythropoietic suppression in the cold-adapted notothenioid lineages through evolutionary modulation of the multi-functional TGF-ß signalling pathway. This trend is more pronounced in the haemoglobin-less icefish, which may pre-emptively hinder the otherwise defective erythroids from production.


Asunto(s)
Evolución Biológica , Eritropoyesis , Perciformes/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Animales , Regiones Antárticas , MicroARNs/genética , Filogenia , Selección Genética , Análisis de Secuencia de ARN , Proteínas de la Superfamilia TGF-beta/genética , Transcriptoma
10.
Nihon Rinsho ; 72(6): 1163-71, 2014 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-25016821

RESUMEN

Marfan syndrome is an autosomal dominant disorder, characterized by tall stature, long arms and legs, ectopia lentis, and aortic aneurysms and dissections. Recent research has revealed that these phenotypes are caused by mutations in fibrillin-1, the major structural component of elastic microfibrils, and the continuing dysregulation of transforming growth factor beta (TGFbeta) signaling is principally considered to be contributing to the pathophysiological background of the disease. Blockade of TGFbeta signaling by angiotensin II receptor antagonism is a novel promising therapeutic option, and thus such large clinical randomized controlled trials are underway. Here, we review the past development, current status and future perspectives in the research field for Marfan syndrome.


Asunto(s)
Síndrome de Marfan , Fibrina/genética , Humanos , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/genética , Síndrome de Marfan/terapia , Proteínas de la Superfamilia TGF-beta/genética
11.
Stem Cell Rev Rep ; 10(2): 280-94, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24395006

RESUMEN

Recent advance in directed differentiation of pancreatic stem cells offers potential to the development of replacement therapy for diabetic patients. However, the existing differentiation protocols are complex, time-consuming, and costly; thus there is a need for alternative protocols. Given the common developmental origins of liver and pancreas, we sought to develop a novel protocol, devoid of growth factors, by using liver stromal cells (LSCs) derived from human fetal liver. We examined the effects of the LSCs on the differentiation of pancreatic progenitor cells (PPCs) into islet-like cell clusters (ICCs). PPCs and LSCs isolated from 1st to 2nd trimester human fetal tissues underwent co-cultures; differentiation and functionality of ICCs were determined by examining expression of critical markers and secretion of insulin. Co-culture with 2nd but not 1st trimester LSCs enhanced ICC differentiation and functionality without the use of exogenous differentiation 'cocktails'. Differential expression profiles of growth factors from 1st versus 2nd trimester fetal liver were compared. Many morphogenic factors were expressed by LSCs, while insulin-like growth factor 1 (IGF1) was identified as one of the key molecules responsible for the ICC differentiation. This is the first report showing that an LSC-induced microenvironment can enhance ICC differentiation and functionality. Further modifications of the stroma microenvironment may offer an alternative, efficient and cost-effective approach to providing islets for transplantation.


Asunto(s)
Diferenciación Celular , Islotes Pancreáticos/citología , Hígado/citología , Células Madre/fisiología , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/terapia , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Desnudos , Células del Estroma/fisiología , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Semin Respir Crit Care Med ; 34(5): 568-80, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24037626

RESUMEN

Tremendous progress has been made in understanding the genetics of heritable pulmonary arterial hypertension (HPAH) since its description in the 1950s. Germline mutations in the gene coding bone morphogenetic receptor type 2 (BMPR2) are detectable in the majority of cases of HPAH, and in a small proportion of cases of idiopathic pulmonary arterial hypertension (IPAH). Recent advancements in gene sequencing methods have facilitated the discovery of additional genes with mutations among those with and without familial PAH (CAV1, KCNK3). HPAH is an autosomal dominant disease characterized by reduced penetrance, variable expressivity, and female predominance. These characteristics suggest that genetic and nongenetic factors modify disease expression, highlighting areas of active investigation. The reduced penetrance makes genetic counseling complex, as the majority of carriers of PAH-related mutations will never be diagnosed with the disease. This issue is increasingly important, as clinical testing for BMPR2 and other mutations is now available for the evaluation of patients and their at-risk kin. The possibilities to avoid mutation transmission, such as the rapidly advancing field of preimplantation genetic testing, highlight the need for all clinicians to understand the genetic features of PAH risk.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipertensión Pulmonar/genética , Receptores de Activinas Tipo II/genética , Antígenos CD/genética , Caveolina 1/genética , Endoglina , Hipertensión Pulmonar Primaria Familiar , Femenino , Interacción Gen-Ambiente , Asesoramiento Genético , Pruebas Genéticas , Mutación de Línea Germinal , Humanos , Hipertensión Pulmonar/epidemiología , Hipertensión Pulmonar/prevención & control , Masculino , Proteínas del Tejido Nervioso/genética , Penetrancia , Canales de Potasio de Dominio Poro en Tándem/genética , Embarazo , Diagnóstico Preimplantación , Receptores de Superficie Celular/genética , Distribución por Sexo , Proteínas de la Superfamilia TGF-beta/genética
13.
Exp Cell Res ; 319(9): 1264-70, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23454603

RESUMEN

Blood vessels are composed of endothelial cells, mural cells (smooth muscle cells and pericytes) and their shared basement membrane. During embryonic development a multitude of signaling components orchestrate the formation of new vessels. The process is highly dependent on correct dosage, spacing and timing of these signaling molecules. As vessels mature some cascades remain active, albeit at very low levels, and may be reactivated upon demand. Members of the Transforming growth factor ß (TGF-ß) protein family are strongly engaged in developmental angiogenesis but are also regulators of vascular integrity in the adult. In humans various genetic alterations within this protein family cause vascular disorders, involving disintegration of vascular integrity. Here we summarize and discuss recent data gathered from conditional and endothelial cell specific genetic loss-of-function of members of the TGF-ß family in the mouse.


Asunto(s)
Proteínas de la Superfamilia TGF-beta/genética , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Eliminación de Gen , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Proteínas de la Superfamilia TGF-beta/metabolismo , Proteínas de la Superfamilia TGF-beta/fisiología
14.
Cardiovasc Pathol ; 22(4): 245-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23261354

RESUMEN

INTRODUCTION: Myxomatous mitral valve is one of the most common heart valves diseases in human and has been well characterized at a functional and morphological level. Diseased valves are thickened as a result of extracellular matrix remodeling and proteoglycans accumulation accompanied by the disruption of the stratified structures of the leaflets. METHODS: Global transcriptomic analysis was used as a start-up to investigate potential pathogenic mechanisms involved in the development of the human idiopathic myxomatous mitral valve, which have been elusive for many years. RESULTS: These prospective analyses have highlighted the potential role of apparently unrelated molecules in myxomatous mitral valve such as members of the transforming growth factor-ß superfamily, aggrecanases of the "a disintegrin and metalloprotease with thrombospondin repeats I" family, and a weakening of the protection against oxidative stress. We have integrated, in this review, recent transcriptomic data from our laboratory [A. Hulin, C.F. Deroanne, C.A. Lambert, B. Dumont, V. Castronovo, J.O. Defraigne, et al. Metallothionein-dependent up-regulation of TGF-beta2 participates in the remodelling of the myxomatous mitral valve. Cardiovasc Res 2012;93:480-489] and from the publication of Sainger et al. [R. Sainger, J.B. Grau, E. Branchetti, P. Poggio, W.F. Seefried, B.C. Field, et al. Human myxomatous mitral valve prolapse: role of bone morphogenetic protein 4 in valvular interstitial cell activation. J Cell Physiol 2012;227:2595-2604] with existing literature and information issued from the study of monogenic syndromes and animal models. CONCLUSION: Understanding cellular alterations and molecular mechanisms involved in myxomatous mitral valve should help at identifying relevant targets for future effective pharmacological therapy to prevent or reduce its progression.


Asunto(s)
Prolapso de la Válvula Mitral/patología , Válvula Mitral/patología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Animales , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica/métodos , Predisposición Genética a la Enfermedad , Humanos , Válvula Mitral/metabolismo , Prolapso de la Válvula Mitral/genética , Prolapso de la Válvula Mitral/metabolismo , Estrés Oxidativo , Fenotipo , Pronóstico , Proteoglicanos/genética , Proteoglicanos/metabolismo , Serotonina/metabolismo , Transducción de Señal , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/metabolismo , Transcriptoma
15.
Int J Dev Biol ; 56(5): 351-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22811269

RESUMEN

Most endoplasmic reticulum (ER)-retained proteins contain a carboxy-terminal signal sequence called the ER retention signal motif such as the Lys-Asp-Glu-Leu (KDEL) motif. Using this molecular mechanism, we developed a new dominant-negative assay, designated the KDEL-tag trap assay, to negatively regulate secretion of disulfide bond-dependent protein dimers, as typified by TGF-beta superfamily proteins. First, we tested this method on the Nodal protein Xnr5, which is a well-studied mesoderm inducer in vertebrates. Tagging of Xnr5 protein with KDEL at the carboxy-terminus effectively blocked the secretion of Xnr5, resulting in complete inhibition of mesoderm induction in Xenopus embryogenesis. Second, we examined the usefulness of the KDEL-tag trap assay on BMPs, which are well-known negative regulators of neural induction and ventralizing factors during early development, and demonstrated that the functions of the BMP family proteins BMP4 and ADMP were blocked by the KDEL-tag trap assay. Moreover, the technical feasibility of the KDEL-tag trap assay was confirmed in a cell culture system using mouse osteoblasts. Taken together, these results suggest that the KDEL-tag trap assay can be adapted to inhibit a variety of plasma membrane or secreted proteins of a multimeric nature.


Asunto(s)
Embrión no Mamífero/metabolismo , Retículo Endoplásmico/metabolismo , Genes Dominantes , Chaperonas Moleculares , Oligopéptidos/metabolismo , Proteínas de la Superfamilia TGF-beta/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Embrión no Mamífero/citología , Ratones , Mutación/genética , Proteína Nodal/metabolismo , Plásmidos , Señales de Clasificación de Proteína , Proteínas de la Superfamilia TGF-beta/genética , Xenopus laevis/metabolismo
16.
Mol Cell Endocrinol ; 359(1-2): 113-20, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21827823

RESUMEN

Inhibins and activins are members of the transforming growth factor (TGFß) superfamily, that includes the TGFßs, inhibins and activins, bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs). The family members are expressed throughout the human body, and are involved in the regulation of a range of important functions. The precise regulation of the TGFß pathways is critical, and mutations of individual molecules or even minor alterations of signalling will have a significant affect on function, that may lead to development of disease or predisposition to the development of disease. The inhibins and activins regulate aspects of the male and female reproductive system, therefore, it is not surprising that most of the diseases associated with abnormalities of the inhibin and activin genes are focused on reproductive disorders and reproductive cancers. In this review, I highlight the role of genetic variants in the development of conditions such as premature ovarian failure, pre-eclampsia, and various reproductive cancers. Given the recent advances in human genetic research, such as genome wide association studies and next generation sequencing, it is likely that inhibins and activins will be shown to play more important roles in a range of human genetic diseases in the future.


Asunto(s)
Activinas/genética , Inhibinas/genética , Mutación , Activinas/metabolismo , Activinas/fisiología , Animales , Neoplasias de las Glándulas Endocrinas/genética , Femenino , Neoplasias de los Genitales Masculinos/genética , Humanos , Inhibinas/metabolismo , Inhibinas/fisiología , Masculino , Preeclampsia/genética , Embarazo , Insuficiencia Ovárica Primaria/genética , Transducción de Señal , Proteínas de la Superfamilia TGF-beta/genética , Gemelos Dicigóticos/genética
17.
Gene ; 472(1-2): 7-17, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21047546

RESUMEN

The gonadal soma-derived factor (GSDF) belongs to the transforming growth factor-ß superfamily and is conserved in teleostean fish species. Gsdf is specifically expressed in the gonads, and gene expression is restricted to the granulosa and Sertoli cells in trout and medaka. The gsdf gene expression is correlated to early testis differentiation in medaka and was shown to stimulate primordial germ cell and spermatogonia proliferation in trout. In the present study, we show that the gsdf gene localizes to a syntenic chromosomal fragment conserved among vertebrates although no gsdf-related gene is detected on the corresponding genomic region in tetrapods. We demonstrate using quantitative RT-PCR that most of the genes localized in the synteny are specifically expressed in medaka gonads. Gsdf is the only gene of the synteny with a much higher expression in the testis compared to the ovary. In contrast, gene expression pattern analysis of the gsdf surrounding genes (nup54, aff1, klhl8, sdad1, and ptpn13) indicates that these genes are preferentially expressed in the female gonads. The tissue distribution of these genes is highly similar in medaka and zebrafish, two teleostean species that have diverged more than 110 million years ago. The cellular localization of these genes was determined in medaka gonads using the whole-mount in situ hybridization technique. We confirm that gsdf gene expression is restricted to Sertoli and granulosa cells in contact with the premeiotic and meiotic cells. The nup54 gene is expressed in spermatocytes and previtellogenic oocytes. Transcripts corresponding to the ovary-specific genes (aff1, klhl8, and sdad1) are detected only in previtellogenic oocytes. No expression was detected in the gonocytes in 10 dpf embryos. In conclusion, we show that the gsdf gene localizes to a syntenic chromosomal fragment harboring evolutionary conserved genes in vertebrates. These genes are preferentially expressed in previtelloogenic oocytes, and thus, they display a different cellular localization compared to that of the gsdf gene indicating that the later gene is not co-regulated. Interestingly, our study identifies new clustered genes that are specifically expressed in previtellogenic oocytes (nup54, aff1, klhl8, sdad1).


Asunto(s)
Proteínas de Peces/genética , Oocitos/metabolismo , Oryzias/genética , Proteínas de la Superfamilia TGF-beta/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Gónadas/metabolismo , Células de la Granulosa/metabolismo , Masculino , Familia de Multigenes , Oocitos/citología , Oryzias/embriología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Testículo/metabolismo , Vitelogeninas/metabolismo
18.
J Biol Chem ; 286(7): 5087-99, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21135108

RESUMEN

The specific functions of the prodomains of TGFß superfamily members are largely unknown. Interactions are known between prodomains of TGFß-1-3 and latent TGFß-binding proteins and between prodomains of BMP-2, -4, -7, and -10 and GDF-5 and fibrillins, raising the possibility that latent TGFß-binding proteins and fibrillins may mediate interactions with all other prodomains of this superfamily. This possibility is tested in this study. Results show that the prodomain of BMP-5 interacts with the N-terminal regions of fibrillin-1 and -2 in a site similar to the binding sites for other bone morphogenetic proteins. However, in contrast, the prodomain of GDF-8 (myostatin) interacts with the glycosaminoglycan side chains of perlecan. The binding site for the GDF-8 prodomain is likely the heparan sulfate chain present on perlecan domain V. These results support and extend the emerging concept that TGFß superfamily prodomains target their growth factor dimers to extracellular matrix macromolecules. In addition, biochemical studies of prodomain·growth factor complexes were performed to identify inactive complexes. For some members of the superfamily, the prodomain is noncovalently associated with its growth factor dimer in an inactive complex; for others, the prodomain·growth factor complex is active, even though the prodomain is noncovalently associated with its growth factor dimer. Results show that the BMP-10 prodomain, in contrast to BMP-4, -5, and -7 prodomains, can inhibit the bioactivity of the BMP-10 growth factor and suggest that the BMP-10 complex is like TGFß and GDF-8 complexes, which can be activated by cleavage of the associated prodomain.


Asunto(s)
Proteínas de la Superfamilia TGF-beta/química , Proteínas de la Superfamilia TGF-beta/metabolismo , Animales , Línea Celular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Proteínas de la Superfamilia TGF-beta/genética
20.
Cytokine Growth Factor Rev ; 20(5-6): 379-88, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19926329

RESUMEN

Smad proteins are intracellular molecules that mediate the canonical signaling cascade of TGFbeta superfamily growth factors. The TGFbeta superfamily comprises two groups of growth factors, BMPs and TGFbetas. Both groups can be further divided into several sub-groups based on sequence homologies and functional similarities. Ligands of the TGFbeta superfamily bind to cell surface receptors to activate Smad proteins in the cytoplasm; then the activated Smad proteins translocate into the nucleus to activate or repress specific target gene transcription. Both groups of growth factors play important roles in skeletal development and regeneration. However, whether these effects reflect signaling through canonical Smad pathways, or other non-canonical signaling pathways in vivo remains a mystery. Moreover, the mechanisms utilized by Smad proteins to initiate nuclear events and their interactions with cytoplasmic proteins are still under intensive investigation. This review will discuss the most recent progress understanding Smad signaling in the context of skeletal development and regeneration.


Asunto(s)
Desarrollo Óseo/genética , Regeneración Ósea/genética , Proteínas Smad/fisiología , Animales , Desarrollo Óseo/fisiología , Regeneración Ósea/fisiología , Condrogénesis/genética , Condrogénesis/fisiología , Humanos , Modelos Biológicos , Osteogénesis/genética , Osteogénesis/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Smad/genética , Proteínas de la Superfamilia TGF-beta/genética , Proteínas de la Superfamilia TGF-beta/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA