Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Thyroid ; 31(3): 420-438, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32777984

RESUMEN

Background: Congenital hypothyroidism due to thyroid dysgenesis is a frequent congenital endocrine disorder for which the molecular mechanisms remain unresolved in the majority of cases. This situation reflects, in part, our still limited knowledge about the mechanisms involved in the early steps of thyroid specification from the endoderm, in particular the extrinsic signaling cues that regulate foregut endoderm patterning. In this study, we used small molecules and genetic zebrafish models to characterize the role of various signaling pathways in thyroid specification. Methods: We treated zebrafish embryos during different developmental periods with small-molecule compounds known to manipulate the activity of Wnt signaling pathway and observed effects in thyroid, endoderm, and cardiovascular development using whole-mount in situ hybridization and transgenic fluorescent reporter models. We used the antisense morpholino (MO) technique to create a zebrafish acardiac model. For thyroid rescue experiments, bone morphogenetic protein (BMP) pathway induction in zebrafish embryos was obtained by manipulation of heat-shock inducible transgenic lines. Results: Combined analyses of thyroid and cardiovascular development revealed that overactivation of Wnt signaling during early development leads to impaired thyroid specification concurrent with severe defects in the cardiac specification. When using a model of MO-induced blockage of cardiomyocyte differentiation, a similar correlation was observed, suggesting that defective signaling between cardiac mesoderm and endodermal thyroid precursors contributes to thyroid specification impairment. Rescue experiments through transient overactivation of BMP signaling could partially restore thyroid specification in models with defective cardiac development. Conclusion: Collectively, our results indicate that BMP signaling is critically required for thyroid cell specification and identify cardiac mesoderm as a likely source of BMP signals.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Hipotiroidismo Congénito/metabolismo , Proteínas del Citoesqueleto/metabolismo , Cardiopatías Congénitas/metabolismo , Miocitos Cardíacos/metabolismo , Disgenesias Tiroideas/metabolismo , Glándula Tiroides/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 4/genética , Hipotiroidismo Congénito/genética , Hipotiroidismo Congénito/patología , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Desarrollo Embrionario , Endodermo/anomalías , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Mesodermo/anomalías , Mesodermo/metabolismo , Morfolinos/genética , Morfolinos/metabolismo , Miocitos Cardíacos/patología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Disgenesias Tiroideas/genética , Disgenesias Tiroideas/patología , Glándula Tiroides/anomalías , Proteínas Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
2.
Toxicology ; 438: 152444, 2020 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-32283119

RESUMEN

As a common birth defect, Cleft palate can be caused by the disturbance during the developmental process of the palatal shelves. The 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) is a well-known environmental teratogenic agent for cleft palate and Aryl hydrocarbon receptor (AhR) pathway can be activated by dioxins. Oct4 as a pluripotent stem cell transcription factor is also involved in the process of embryonic development. The AHR and retinoid receptors have cross-talk at CYP1A1 (cytochrome P450, family 1, subfamily A, polypeptide 1) promoter. There are also bidirectional talk between AhR and Oct4. In this study, we used C57/BL6 N mice and TCDD (64 µg/Kg body weight) to establish a model of fetal cleft palate to observe the effects of dioxin on fetal mesenchymal proliferation and apoptosis, and explore the role of Oct4 in inducing cleft palate. The results showed that dioxin inhibited mesenchymal proliferation and promoted apoptosis. In addition, dioxin inhibited Oct4 expression, and preliminary data suggest that hypermethylation of the Oct4 promoter may be a putative mechanism, suggesting that TCDD might induce cleft palate by inhibiting the proliferation of palatal mesenchymal cells mediated by Oct4.


Asunto(s)
Proliferación Celular , Fisura del Paladar/metabolismo , Mesodermo/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Hueso Paladar/metabolismo , Dibenzodioxinas Policloradas , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Fisura del Paladar/inducido químicamente , Fisura del Paladar/patología , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Masculino , Mesodermo/anomalías , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , Hueso Paladar/anomalías , Embarazo , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal
3.
Int J Mol Sci ; 20(7)2019 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-30974877

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects derived from abnormalities in renal differentiation during embryogenesis. CAKUT is the major cause of end-stage renal disease and chronic kidney diseases in children, but its genetic causes remain largely unresolved. Here we discuss advances in the understanding of how mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) activity contributes to the regulation of ureteric bud branching morphogenesis, which dictates the final size, shape, and nephron number of the kidney. Recent studies also demonstrate that the MAPK/ERK pathway is directly involved in nephrogenesis, regulating both the maintenance and differentiation of the nephrogenic mesenchyme. Interestingly, aberrant MAPK/ERK signaling is linked to many cancers, and recent studies suggest it also plays a role in the most common pediatric renal cancer, Wilms' tumor.


Asunto(s)
Diferenciación Celular , Riñón , Sistema de Señalización de MAP Quinasas , Mesodermo , Organogénesis , Animales , Humanos , Riñón/anomalías , Riñón/embriología , Mesodermo/anomalías , Mesodermo/embriología
4.
J Int Adv Otol ; 15(2): 317-320, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30431015

RESUMEN

Axial mesodermal dysplasia complex (AMDC) is a combination of multiple congenital malformations arising due to the mesodermal cell migration, neural tube fusion, and rhombencephalon segmentation. Here, we present the imaging findings of a 15-year-old boy with AMDC who has bilateral inner ear malformations associated with a vestibulocochlear nerve extending to Meckel cave, cystic lesion in prepontine cisterna, cervical vertebral segmentation anomalies, and maxillar bone anomalies.


Asunto(s)
Anomalías Múltiples/patología , Oído Interno/anomalías , Mesodermo/anomalías , Nervio Vestibulococlear/anomalías , Adolescente , Vértebras Cervicales/anomalías , Humanos , Imagen por Resonancia Magnética , Masculino , Tomografía Computarizada por Rayos X
5.
Biol Reprod ; 100(1): 49-60, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010727

RESUMEN

Wnt4 and Wnt5a have well-established roles in the embryonic development of the female reproductive tract, as well as in implantation, decidualization, and ovarian function in adult mice. Although these roles appear to overlap, whether Wnt5a and Wnt4 are functionally redundant in these tissues has not been determined. We addressed this by concomitantly inactivating Wnt4 and Wnt5a in the Müllerian mesenchyme and in ovarian granulosa cells by crossing mice bearing floxed alleles to the Amhr2cre strain. Whereas fertility was reduced by ∼50% in Wnt4flox/flox; Amhr2cre/+ and Wnt5aflox/flox; Amhr2cre/+ females, Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice were either nearly or completely sterile. Loss of fertility was not due to an ovarian defect, as serum ovarian hormone levels, follicle counts, and ovulation rates were comparable to controls. Conversely, the uterus was abnormal in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice, with thin myometrial and stromal layers, frequent fibrosis and a >90% reduction in numbers of uterine glands, suggesting redundant or additive roles of Wnt4 and Wnt5a in uterine adenogenesis. Loss of fertility in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice was attributed to defects in decidualization, implantation, and placental development, the severity of which were proportional to the extent of gland loss. Furthermore, a third of Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ females had a partial agenesis of Müllerian duct-derived structures, but with normal oviducts and ovaries. Together, our results suggest that Wnt4 and Wnt5a play redundant roles in the development of the female reproductive tract, and may provide insight into the etiology of certain cases of Müllerian agenesis in women.


Asunto(s)
Conductos Paramesonéfricos/anomalías , Conductos Paramesonéfricos/metabolismo , Anomalías Urogenitales/genética , Útero/anomalías , Proteína Wnt-5a/genética , Proteína Wnt4/genética , Animales , Femenino , Eliminación de Gen , Infertilidad Femenina/embriología , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Masculino , Mesodermo/anomalías , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Ratones Transgénicos , Conductos Paramesonéfricos/patología , Miometrio/anomalías , Miometrio/metabolismo , Embarazo , Anomalías Urogenitales/metabolismo , Anomalías Urogenitales/patología , Útero/metabolismo , Proteína Wnt-5a/metabolismo , Proteína Wnt4/metabolismo
6.
J Biol Chem ; 292(34): 14165-14175, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28687631

RESUMEN

Circadian clock and Smad2/3/4-mediated Nodal signaling regulate multiple physiological and pathological processes. However, it remains unknown whether Clock directly cross-talks with Nodal signaling and how this would regulate embryonic development. Here we show that Clock1a coordinated mesoderm development and primitive hematopoiesis in zebrafish embryos by directly up-regulating Nodal-Smad3 signaling. We found that Clock1a is expressed both maternally and zygotically throughout early zebrafish development. We also noted that Clock1a alterations produce embryonic defects with shortened body length, lack of the ventral tail fin, or partial defect of the eyes. Clock1a regulates the expression of the mesodermal markers ntl, gsc, and eve1 and of the hematopoietic markers scl, lmo2, and fli1a Biochemical analyses revealed that Clock1a stimulates Nodal signaling by increasing expression of Smad2/3/4. Mechanistically, Clock1a activates the smad3a promoter via its E-box1 element (CAGATG). Taken together, these findings provide mechanistic insight into the role of Clock1a in the regulation of mesoderm development and primitive hematopoiesis via modulation of Nodal-Smad3 signaling and indicate that Smad3a is directly controlled by the circadian clock in zebrafish.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario , Mesodermo/metabolismo , Proteína Nodal/agonistas , Transducción de Señal , Proteína smad3/agonistas , Proteínas de Pez Cebra/agonistas , Pez Cebra , Animales , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Embrión no Mamífero/anomalías , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hematopoyesis/efectos de los fármacos , Humanos , Hibridación in Situ , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mesodermo/anomalías , Mesodermo/citología , Mesodermo/efectos de los fármacos , Microinyecciones , Microscopía Fluorescente , Morfolinos/farmacología , Mutación , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética , Proteína smad3/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Sci Rep ; 7: 42170, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176844

RESUMEN

Previously genome-wide association methods in patients with classic bladder exstrophy (CBE) found association with ISL1, a master control gene expressed in pericloacal mesenchyme. This study sought to further explore the genetics in a larger set of patients following-up on the most promising genomic regions previously reported. Genotypes of 12 markers obtained from 268 CBE patients of Australian, British, German Italian, Spanish and Swedish origin and 1,354 ethnically matched controls and from 92 CBE case-parent trios from North America were analysed. Only marker rs6874700 at the ISL1 locus showed association (p = 2.22 × 10-08). A meta-analysis of rs6874700 of our previous and present study showed a p value of 9.2 × 10-19. Developmental biology models were used to clarify the location of ISL1 activity in the forming urinary tract. Genetic lineage analysis of Isl1-expressing cells by the lineage tracer mouse model showed Isl1-expressing cells in the urinary tract of mouse embryos at E10.5 and distributed in the bladder at E15.5. Expression of isl1 in zebrafish larvae staged 48 hpf was detected in a small region of the developing pronephros. Our study supports ISL1 as a major susceptibility gene for CBE and as a regulator of urinary tract development.


Asunto(s)
Extrofia de la Vejiga/genética , Predisposición Genética a la Enfermedad , Proteínas con Homeodominio LIM/genética , Mesodermo/metabolismo , Organogénesis/genética , Factores de Transcripción/genética , Sistema Urinario/metabolismo , Animales , Extrofia de la Vejiga/metabolismo , Extrofia de la Vejiga/patología , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas con Homeodominio LIM/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Mesodermo/anomalías , Mesodermo/crecimiento & desarrollo , Ratones , Polimorfismo de Nucleótido Simple , Pronefro/crecimiento & desarrollo , Pronefro/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo , Sistema Urinario/anomalías , Sistema Urinario/crecimiento & desarrollo , Pez Cebra
9.
BMC Biol ; 14: 19, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26984772

RESUMEN

BACKGROUND: Epithelial-mesenchymal cross talk is centerpiece in the development of many branched organs, including the lungs. The embryonic lung mesoderm provides instructional information not only for lung architectural development, but also for patterning, commitment and differentiation of its many highly specialized cell types. The mesoderm also serves as a reservoir of progenitors for generation of differentiated mesenchymal cell types that include αSMA-expressing fibroblasts, lipofibroblasts, endothelial cells and others. Transforming Growth Factor ß (TGFß) is a key signaling pathway in epithelial-mesenchymal cross talk. Using a cre-loxP approach we have elucidated the role of the TGFß type I receptor tyrosine kinase, ALK5, in epithelial-mesenchymal cross talk during lung morphogenesis. RESULTS: Targeted early inactivation of Alk5 in mesodermal progenitors caused abnormal development and maturation of the lung that included reduced physical size of the sub-mesothelial mesoderm, an established source of specific mesodermal progenitors. Abrogation of mesodermal ALK5-mediated signaling also inhibited differentiation of cell populations in the epithelial and endothelial lineages. Importantly, Alk5 mutant lungs contained a reduced number of αSMA(pos) cells and correspondingly increased lipofibroblasts. Elucidation of the underlying mechanisms revealed that through direct and indirect modulation of target signaling pathways and transcription factors, including PDGFRα, PPARγ, PRRX1, and ZFP423, ALK5-mediated TGFß controls a process that regulates the commitment and differentiation of αSMA(pos) versus lipofibroblast cell populations during lung development. CONCLUSION: ALK5-mediated TGFß signaling controls an early pathway that regulates the commitment and differentiation of αSMA(pos) versus LIF cell lineages during lung development.


Asunto(s)
Pulmón/citología , Pulmón/embriología , Mesodermo/citología , Mesodermo/embriología , Miofibroblastos/citología , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Células Madre/citología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Pulmón/anomalías , Pulmón/metabolismo , Mesodermo/anomalías , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Músculo Liso/anomalías , Músculo Liso/citología , Músculo Liso/embriología , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Células Madre/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Dev Biol ; 409(1): 106-113, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26485363

RESUMEN

The developmental process through which the cloaca transforms from one hollow structure to two separated urinary and digestive outlets remains controversial and speculative. Here, we use high-resolution episcopic microscopy to examine a comprehensive series of normal and mutant mouse cloaca in which the detailed 3-dimensional (3-D) morphological features are illuminated throughout the development. We provide evidence that the dorsal peri-cloacal mesenchyme (dPCM) remains stationary while other surrounding tissues grow towards it. This causes dramatic changes of spatial relationship among caudal structures and morphological transformation of the cloaca. The 3-D characterizations of Dkk1 mutants reveal a hyperplastic defect of dPCM, which leads to a significant anterior shift of the caudal boundary of the cloaca, premature occlusion of the cloaca and, imperforate anus phenotype. Conversely, Shh knockout causes a severe hypoplastic defect of cloaca mesenchyme including dPCM and persistent cloaca. Collectively, these findings suggest that formation of the dPCM is critical for cloacal morphogenesis and furthermore, growth and movement of the mesenchymal tissues towards the dPCM lead to the cloaca occlusion and separation of the urinary and digestive outlets.


Asunto(s)
Cloaca/anatomía & histología , Cloaca/embriología , Mamíferos/embriología , Microscopía/métodos , Morfogénesis , Canal Anal/anomalías , Canal Anal/embriología , Canal Anal/patología , Animales , Malformaciones Anorrectales , Ano Imperforado/embriología , Ano Imperforado/patología , Imagenología Tridimensional , Mesodermo/anomalías , Mesodermo/embriología , Mesodermo/patología , Ratones Endogámicos C57BL , Recto/anomalías , Recto/embriología , Recto/patología , Anomalías Urogenitales/embriología , Anomalías Urogenitales/patología
12.
BMC Biol ; 13: 42, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-26092405

RESUMEN

BACKGROUND: Adenomatous polyposis coli (Apc) is a tumor suppressor that inhibits Wnt/Ctnnb1. Mutations of Apc will not only lead to familial adenomatous polyposis with associated epithelial lesions, but will also cause aggressive fibromatosis in mesenchymal cells. However, the roles of Apc in regulating mesenchymal cell biology and organogenesis during development are unknown. RESULTS: We have specifically deleted the Apc gene in lung mesenchymal cells during early lung development in mice. Loss of Apc function resulted in immediate mesenchymal cell hyperproliferation through abnormal activation of Wnt/Ctnnb1, followed by a subsequent inhibition of cell proliferation due to cell cycle arrest at G0/G1, which was caused by a mechanism independent of Wnt/Ctnnb1. Meanwhile, abrogation of Apc also disrupted lung mesenchymal cell differentiation, including decreased airway and vascular smooth muscle cells, the presence of Sox9-positive mesenchymal cells in the peripheral lung, and excessive versican production. Moreover, lung epithelial branching morphogenesis was drastically inhibited due to disrupted Bmp4-Fgf10 morphogen production and regulation in surrounding lung mesenchyme. Lastly, lung mesenchyme-specific Apc conditional knockout also resulted in altered lung vasculogenesis and disrupted pulmonary vascular continuity through a paracrine mechanism, leading to massive pulmonary hemorrhage and lethality at mid-gestation when the pulmonary circulation should have started. CONCLUSIONS: Our study suggests that Apc in lung mesenchyme plays central roles in coordinating the proper development of several quite different cellular compartments including lung epithelial branching and pulmonary vascular circulation during lung organogenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Pulmón/anomalías , Pulmón/crecimiento & desarrollo , Mesodermo/citología , Animales , Diferenciación Celular , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Mesodermo/anomalías , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
13.
Am J Physiol Renal Physiol ; 308(8): F888-98, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25656370

RESUMEN

While urothelial signals, including sonic hedgehog (Shh), drive bladder mesenchyme differentiation, it is unclear which pathways within the mesenchyme are critical for its development. Studies have shown that fibroblast growth factor receptor (Fgfr)2 is necessary for kidney and ureter mesenchymal development. The objective of the present study was to determine the role of Fgfr2 in the bladder mesenchyme. We used Tbx18cre mice to delete Fgfr2 in the bladder mesenchyme (Fgfr2(BM-/-)). We performed three-dimensional reconstructions, quantitative real-time PCR, in situ hybridization, immunolabeling, ELISAs, immunoblot analysis, void stain on paper, ex vivo bladder sheet assays, and in vivo decerebrated cystometry. Compared with control bladders, embryonic day 16.5 (E16.5) Fgfr2(BM-/-) bladders had thin muscle layers with less α-smooth muscle actin and thickened lamina propria with increased collagen type Ia and IIIa that intruded into the muscle. The reciprocal changes in mutant layer thicknesses appeared partly due to a cell fate switch. From postnatal days 1 to 30, Fgfr2(BM-/-) bladders demonstrated progressive muscle loss and increased collagen expression. Postnatal Fgfr2(BM-/-) bladder sheets exhibited decreased agonist-mediated contractility and increased passive stretch tension versus control bladder sheets. Cystometry revealed high baseline and threshold pressures and shortened intercontractile intervals in Fgfr2(BM-/-) versus control bladders. Mechanistically, whereas Shh expression appeared normal, mRNA and protein readouts of hedgehog activity were increased in E16.5 Fgfr2(BM-/-) versus control bladders. Moreover, E16.5 Fgfr2(BM-/-) bladders exhibited higher levels of Cdo and Boc, hedgehog coreceptors that enhance sensitivity to Shh, compared with control bladders. In conclusion, loss of Fgfr2 in the bladder mesenchyme leads to abnormal bladder morphology and decreased compliance and contractility.


Asunto(s)
Tipificación del Cuerpo , Mesodermo/metabolismo , Músculo Liso/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Vejiga Urinaria/metabolismo , Animales , Apoptosis , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Adaptabilidad , Fibrosis , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Proteínas Hedgehog/metabolismo , Inmunoglobulina G/metabolismo , Masculino , Mesodermo/anomalías , Ratones Noqueados , Contracción Muscular , Músculo Liso/anomalías , Músculo Liso/fisiopatología , Tamaño de los Órganos , Fenotipo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Vejiga Urinaria/anomalías , Vejiga Urinaria/fisiopatología , Urodinámica
14.
Pediatr Dev Pathol ; 18(2): 146-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25625757

RESUMEN

We report a dichorionic twin gestation with diffuse placental mesenchymal dysplasia (PMD) and androgenetic biparental mosaicism (ABM) involving one twin's placenta with complete absence of fetal development for that twin. To our knowledge, this is the 1st reported case of PMD without fetal development. We discuss the gross, histologic, and genetic hallmarks of PMD and the spectrum of variability depending on degree and distribution of ABM.


Asunto(s)
Mesodermo/anomalías , Mosaicismo , Enfermedades Placentarias/genética , Placenta/anomalías , Embarazo Gemelar , Gemelos Dicigóticos/genética , Adulto , Biopsia , Femenino , Fertilización In Vitro , Predisposición Genética a la Enfermedad , Edad Gestacional , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Humanos , Hibridación Fluorescente in Situ , Masculino , Fenotipo , Enfermedades Placentarias/diagnóstico , Valor Predictivo de las Pruebas , Embarazo , Factores de Riesgo , Ultrasonografía Prenatal
15.
Arch Pathol Lab Med ; 138(9): 1247-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25171710

RESUMEN

Placental mesenchymal dysplasia is a rare placental lesion characterized by stem villous cystic dilation and vesicle formation, placentomegaly, and vascular abnormalities. It can be associated with growth restriction, stillbirth, Beckwith-Wiedemann syndrome, and some chromosomal abnormalities, and needs to be distinguished from its main differential diagnosis, hydatidiform mole.


Asunto(s)
Mesodermo/anomalías , Enfermedades Placentarias/diagnóstico , Enfermedades Placentarias/patología , Placenta/anomalías , Síndrome de Beckwith-Wiedemann , Aberraciones Cromosómicas , Diagnóstico Diferencial , Femenino , Retardo del Crecimiento Fetal , Humanos , Mola Hidatiforme/diagnóstico , Mesodermo/patología , Placenta/patología , Enfermedades Placentarias/epidemiología , Embarazo
16.
Kobe J Med Sci ; 60(1): E1-4, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-25011636

RESUMEN

Placental mesenchymal dysplasia (PMD) is a rare condition presenting with enlarged, multicystic placenta like molar changes. Although PMD usually features a normal fetus and the pregnancy often extends into the third trimester, PMD is clinically significant lesion with high rates of FGR, IUFD, and is associated with Beckwith-Wiedemann syndrome (BWS). We report a 30-year old woman at her first pregnancy with intrauterine sudden death at 31 weeks of gestation. The vesicular lesion in her uterus was detected at 10 weeks on ultrasound. The fetus was normal size without any anomaly on ultrasound and normal trophoblastic vascularization by Doppler study during the pregnancy. As the pregnancy advanced, the vesicular lesion decreased in size and no fetal abnormalities were detected. At 28 weeks of gestation an ultrasound detected dilated periumbilical chorionic vessels. We didn't detect severe FGR or abnormal trophoblastic vascularization. At 31 weeks of gestation an intrauterine sudden death of a normal-sized fetus without any anomaly occurred. The placenta was enlarged, and microscopic morphology confirmed a diagnosis of PMD. The chorionic vessels were cirsoid, dilated and tortuous. We determined the rupture of expanded periumbilical chorionic vessels led to fetal death.


Asunto(s)
Muerte Perinatal/etiología , Placenta/anomalías , Adulto , Vellosidades Coriónicas/anomalías , Vellosidades Coriónicas/irrigación sanguínea , Femenino , Edad Gestacional , Humanos , Mesodermo/anomalías , Mesodermo/diagnóstico por imagen , Placenta/irrigación sanguínea , Placenta/diagnóstico por imagen , Embarazo , Rotura Espontánea , Ultrasonografía Prenatal
17.
Am J Physiol Renal Physiol ; 306(7): F764-72, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24500693

RESUMEN

MicroRNAs (miRNAs) are small, noncoding regulatory RNAs that act as posttranscriptional repressors by binding to the 3'-untranslated region (3'-UTR) of target genes. They require processing by Dicer, an RNase III enzyme, to become mature regulatory RNAs. Previous work from our laboratory revealed critical roles for miRNAs in nephron progenitors at midgestation (Ho J, Pandey P, Schatton T, Sims-Lucas S, Khalid M, Frank MH, Hartwig S, Kreidberg JA. J Am Soc Nephrol 22: 1053-1063, 2011). To interrogate roles for miRNAs in the early metanephric mesenchyme, which gives rise to nephron progenitors as well as the renal stroma during kidney development, we conditionally ablated Dicer function in this lineage. Despite normal ureteric bud outgrowth and condensation of the metanephric mesenchyme to form nephron progenitors, early loss of miRNAs in the metanephric mesenchyme resulted in severe renal dysgenesis. Nephron progenitors are initially correctly specified in the mutant kidneys, with normal expression of several transcription factors known to be critical in progenitors, including Six2, Pax2, Sall1, and Wt1. However, there is premature loss of the nephron progenitor marker Cited1, marked apoptosis, and increased expression of the proapoptotic protein Bim shortly after the initial inductive events in early kidney development. Subsequently, there is a failure in ureteric bud branching and nephron progenitor differentiation. Taken together, our data demonstrate a previously undetermined requirement for miRNAs during early kidney organogenesis and indicate a crucial role for miRNAs in regulating the survival of this lineage.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Células Madre Embrionarias/enzimología , Riñón/enzimología , Mesodermo/enzimología , Ribonucleasa III/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Riñón/anomalías , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/anomalías , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Nefronas/anomalías , Nefronas/enzimología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organogénesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ribonucleasa III/deficiencia , Ribonucleasa III/genética , Transactivadores/genética , Transactivadores/metabolismo , Uréter/anomalías , Uréter/enzimología
18.
Dis Model Mech ; 6(3): 755-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23471918

RESUMEN

The ZIC transcription factors are key mediators of embryonic development and ZIC3 is the gene most commonly associated with situs defects (heterotaxy) in humans. Half of patient ZIC3 mutations introduce a premature termination codon (PTC). In vivo, PTC-containing transcripts might be targeted for nonsense-mediated decay (NMD). NMD efficiency is known to vary greatly between transcripts, tissues and individuals and it is possible that differences in survival of PTC-containing transcripts partially explain the striking phenotypic variability that characterizes ZIC3-associated congenital defects. For example, the PTC-containing transcripts might encode a C-terminally truncated protein that retains partial function or that dominantly interferes with other ZIC family members. Here we describe the katun (Ka) mouse mutant, which harbours a mutation in the Zic3 gene that results in a PTC. At the time of axis formation there is no discernible decrease in this PTC-containing transcript in vivo, indicating that the mammalian Zic3 transcript is relatively insensitive to NMD, prompting the need to re-examine the molecular function of the truncated proteins predicted from human studies and to determine whether the N-terminal portion of ZIC3 possesses dominant-negative capabilities. A combination of in vitro studies and analysis of the Ka phenotype indicate that it is a null allele of Zic3 and that the N-terminal portion of ZIC3 does not encode a dominant-negative molecule. Heterotaxy in patients with PTC-containing ZIC3 transcripts probably arises due to loss of ZIC3 function alone.


Asunto(s)
Codón sin Sentido/genética , Síndrome de Heterotaxia/embriología , Síndrome de Heterotaxia/genética , Proteínas de Homeodominio/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido/genética , Factores de Transcripción/metabolismo , Alelos , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Difusión , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Endodermo/anomalías , Endodermo/embriología , Gastrulación/genética , Proteínas de Homeodominio/genética , Humanos , Mesodermo/anomalías , Mesodermo/embriología , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Organogénesis/genética , Estabilidad Proteica , Sitios de Empalme de ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Transcripción Genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA