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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38546045

RESUMEN

The primary cilium decorates most eukaryotic cells and regulates tissue morphogenesis and maintenance. Structural or functional defects of primary cilium result in ciliopathies, congenital human disorders affecting multiple organs. Pathogenic variants in the ciliogenesis and planar cell polarity effectors (CPLANE) genes FUZZY, INTU and WDPCP disturb ciliogenesis, causing severe ciliopathies in humans and mice. Here, we show that the loss of Fuzzy in mice results in defects of primary cilia, accompanied by increased RhoA activity and excessive actin polymerization at the basal body. We discovered that, mechanistically, Fuzzy interacts with and recruits the negative actin regulator ARHGAP35 (also known as p190A RhoGAP) to the basal body. We identified genetic interactions between the two genes and found that a mutant ArhGAP35 allele increases the severity of phenotypic defects observed in Fuzzy-/- mice. Based on our findings, we propose that Fuzzy regulates ciliogenesis by recruiting ARHGAP35 to the basal body, where the latter likely restricts actin polymerization and modifies the actin network. Our study identifies a mechanism whereby CPLANE proteins control both actin polymerization and primary cilium formation.


Asunto(s)
Actinas , Ciliopatías , Proteínas Activadoras de GTPasa , Ratones , Humanos , Animales , Actinas/metabolismo , Cilios/metabolismo , Polimerizacion
2.
Nucleic Acids Res ; 51(14): 7314-7329, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37395395

RESUMEN

ZMYM2 is a transcriptional repressor whose role in development is largely unexplored. We found that Zmym2-/- mice show embryonic lethality by E10.5. Molecular characterization of Zmym2-/- embryos revealed two distinct defects. First, they fail to undergo DNA methylation and silencing of germline gene promoters, resulting in widespread upregulation of germline genes. Second, they fail to methylate and silence the evolutionarily youngest and most active LINE element subclasses in mice. Zmym2-/- embryos show ubiquitous overexpression of LINE-1 protein as well as aberrant expression of transposon-gene fusion transcripts. ZMYM2 homes to sites of PRC1.6 and TRIM28 complex binding, mediating repression of germline genes and transposons respectively. In the absence of ZMYM2, hypermethylation of histone 3 lysine 4 occurs at target sites, creating a chromatin landscape unfavourable for establishment of DNA methylation. ZMYM2-/- human embryonic stem cells also show aberrant upregulation and demethylation of young LINE elements, indicating a conserved role in repression of active transposons. ZMYM2 is thus an important new factor in DNA methylation patterning in early embryonic development.


Asunto(s)
Metilación de ADN , Animales , Humanos , Ratones , Cromatina/genética , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario/genética , Células Germinativas/metabolismo , Histonas/genética , Histonas/metabolismo , Factores de Transcripción/metabolismo
3.
Hum Mol Genet ; 28(10): 1671-1681, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30649340

RESUMEN

Hand-Foot-Genital syndrome is a rare condition caused by mutations in the HOXA13 gene and characterized by limb malformations and urogenital defects. While the role of Hoxa13 in limb development has been extensively studied, its function during the development of the urogenital system remains elusive mostly due to the embryonic lethality of Hoxa13 homozygous mutant mice. Using a conditional inactivation strategy, we show that mouse fetuses lacking Hoxa13 function develop megaureters, hydronephrosis and malformations of the uterus, reminiscent of the defects characterizing patients with Hand-Foot-Genital syndrome. Our analysis reveals that Hoxa13 plays a critical role in Müllerian ducts fusion and in ureter remodeling by regulating the elimination of the caudal common nephric duct, eventually preventing the separation from the nephric duct. Our data also reveal a specific role for Hoxa13 in the urogenital sinus, which is in part mediated by Gata3, as well as Hoxa13 requirement for the proper organization of the ureter. Finally, we provide evidence that Hoxa13 provides positional and temporal cues during the development of the lower urogenital system, a sine qua non condition for the proper function of the urinary system.


Asunto(s)
Anomalías Múltiples/genética , Deformidades Congénitas del Pie/genética , Factor de Transcripción GATA3/genética , Deformidades Congénitas de la Mano/genética , Proteínas de Homeodominio/genética , Anomalías Urogenitales/genética , Sistema Urogenital/fisiopatología , Anomalías Múltiples/fisiopatología , Animales , Extremidades/crecimiento & desarrollo , Extremidades/fisiopatología , Deformidades Congénitas del Pie/fisiopatología , Deformidades Congénitas de la Mano/fisiopatología , Humanos , Riñón/anomalías , Riñón/patología , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/fisiopatología , Ratones , Conductos Paramesonéfricos/fisiopatología , Mutación , Uréter/anomalías , Uréter/fisiopatología , Anomalías Urogenitales/fisiopatología , Sistema Urogenital/crecimiento & desarrollo
4.
Development ; 145(20)2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30348673

RESUMEN

The GATA family of transcription factors is of crucial importance during embryonic development, playing complex and widespread roles in cell fate decisions and tissue morphogenesis. GATA proteins are essential for the development of tissues derived from all three germ layers, including the skin, brain, gonads, liver, hematopoietic, cardiovascular and urogenital systems. The crucial activity of GATA factors is underscored by the fact that inactivating mutations in most GATA members lead to embryonic lethality in mouse models and are often associated with developmental diseases in humans. In this Primer, we discuss the unique and redundant functions of GATA proteins in tissue morphogenesis, with an emphasis on their regulation of lineage specification and early organogenesis.


Asunto(s)
Enfermedad , Factores de Transcripción GATA/metabolismo , Crecimiento y Desarrollo , Animales , Diferenciación Celular , Humanos , Organogénesis , Transcripción Genética
5.
Development ; 144(16): 2982-2993, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28705896

RESUMEN

Caudal somites are generated from a pool of progenitor cells located in the tailbud region. These progenitor cells form the presomitic mesoderm that gradually differentiates into somites under the action of the segmentation clock. The signals responsible for tailbud mesoderm progenitor pool maintenance during axial elongation are still elusive. Here, we show that Bmp signaling is sufficient to activate the entire mesoderm progenitor gene signature in primary cultures of caudal mesoderm cells. Bmp signaling acts through the key regulatory genes brachyury (T) and Nkx1-2 and contributes to the activation of several other regulators of the mesoderm progenitor gene network. In the absence of Bmp signaling, tailbud mesoderm progenitor cells acquire aberrant gene expression signatures of the heart, blood, muscle and skeletal embryonic lineages. Treatment of embryos with the Bmp inhibitor noggin confirmed the requirement for Bmp signaling for normal T expression and the prevention of abnormal lineage marker activation. Together, these results identify Bmp signaling as a non-cell-autonomous signal necessary for mesoderm progenitor cell homeostasis.


Asunto(s)
Mesodermo/citología , Mesodermo/embriología , Células Madre/metabolismo , Cola (estructura animal)/citología , Cola (estructura animal)/embriología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Células Madre/citología , Cola (estructura animal)/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(43): E9016-E9025, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073098

RESUMEN

The elimination of unwanted cells by apoptosis is necessary for tissue morphogenesis. However, the cellular control of morphogenetic apoptosis is poorly understood, notably the modulation of cell sensitivity to apoptotic stimuli. Ureter maturation, the process by which the ureter is displaced to the bladder wall, represents an exquisite example of morphogenetic apoptosis, requiring the receptor protein tyrosine phosphatases (RPTPs): LAR and RPTPσ. Here we show that LAR-RPTPs act through cellular inhibitor of apoptosis protein 1 (cIAP1) to modulate caspase 3,7-mediated ureter maturation. Pharmacologic or genetic inactivation of cIAP1 reverts the apoptotic deficit of LAR-RPTP-deficient embryos. Moreover, Birc2 (cIAP1) inactivation generates excessive apoptosis leading to vesicoureteral reflux in newborns, which underscores the importance of apoptotic modulation during urinary tract morphogenesis. We finally demonstrate that LAR-RPTP deficiency increases cIAP1 stability during apoptotic cell death. Together these results identify a mode of cIAP1 regulation playing a critical role in the cellular response to apoptotic pathway activation in the embryo.


Asunto(s)
Apoptosis/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Uréter/embriología , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Células Cultivadas , Embrión de Mamíferos , Fibroblastos/fisiología , Proteínas Inhibidoras de la Apoptosis/genética , Ratones , Ratones Noqueados , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas , Uréter/metabolismo
7.
PLoS Genet ; 12(2): e1005785, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26859289

RESUMEN

Rho family GTPases act as molecular switches regulating actin cytoskeleton dynamics. Attenuation of their signaling capacity is provided by GTPase-activating proteins (GAPs), including p190A, that promote the intrinsic GTPase activity of Rho proteins. In the current study we have performed a small-scale ENU mutagenesis screen and identified a novel loss of function allele of the p190A gene Arhgap35, which introduces a Leu1396 to Gln substitution in the GAP domain. This results in decreased GAP activity for the prototypical Rho-family members, RhoA and Rac1, likely due to disrupted ordering of the Rho binding surface. Consequently, Arhgap35-deficient animals exhibit hypoplastic and glomerulocystic kidneys. Investigation into the cystic phenotype shows that p190A is required for appropriate primary cilium formation in renal nephrons. P190A specifically localizes to the base of the cilia to permit axoneme elongation, which requires a functional GAP domain. Pharmacological manipulations further reveal that inhibition of either Rho kinase (ROCK) or F-actin polymerization is able to rescue the ciliogenesis defects observed upon loss of p190A activity. We propose a model in which p190A acts as a modulator of Rho GTPases in a localized area around the cilia to permit the dynamic actin rearrangement required for cilia elongation. Together, our results establish an unexpected link between Rho GTPase regulation, ciliogenesis and glomerulocystic kidney disease.


Asunto(s)
Cilios/metabolismo , Proteínas Activadoras de GTPasa/genética , Enfermedades Renales Quísticas/genética , Glomérulos Renales/patología , Organogénesis , Mutación Puntual/genética , Proteínas Represoras/genética , Actinas/metabolismo , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Citoesqueleto/metabolismo , Embrión de Mamíferos/citología , Etilnitrosourea , Femenino , Fibroblastos/metabolismo , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Enfermedades Renales Quísticas/patología , Glomérulos Renales/metabolismo , Túbulos Renales/anomalías , Túbulos Renales/patología , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Defectos del Tubo Neural/patología , Fenotipo , Estructura Terciaria de Proteína , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados
8.
Dev Dyn ; 247(11): 1186-1198, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30295986

RESUMEN

BACKGROUND: The tissue-specific transcriptional programs during normal development require tight control by distal cis-regulatory elements, such as enhancers, with specific DNA sequences recognized by transcription factors, coactivators, and chromatin remodeling enzymes. Gata3 is a sequence-specific DNA-binding transcription factor that regulates formation of multiple tissues and organs, including inner ear, lens, mammary gland, T-cells, urogenital system, and thyroid gland. In the eye, Gata3 has a highly restricted expression domain in the posterior part of the lens vesicle; however, the underlying regulatory mechanisms are unknown. RESULTS: Here we describe the identification of a novel bipartite Gata3 lens-specific enhancer located ∼18 kb upstream from its transcriptional start site. We also found that a 5-kb Gata3 promoter possesses low activity in the lens. The bipartite enhancer contains arrays of AP-1, Ets-, and Smad1/5-binding sites as well as binding sites for lens-associated DNA-binding factors. Transient transfection studies of the promoter with the bipartite enhancer showed enhanced activation by BMP4 and FGF2. CONCLUSIONS: These studies identify a novel distal enhancer of Gata3 with high activity in lens and indicate that BMP and FGF signaling can up-regulate expression of Gata3 in differentiating lens fiber cells through the identified Gata3 enhancer and promoter elements. Developmental Dynamics 247:1186-1198, 2018. © 2018 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Asunto(s)
Elementos de Facilitación Genéticos , Factor de Transcripción GATA3/genética , Cristalino/embriología , Animales , Sitios de Unión , Proteína Morfogenética Ósea 4/fisiología , Proteínas de Unión al ADN , Factor 2 de Crecimiento de Fibroblastos/fisiología , Factor de Transcripción GATA3/química , Factor de Transcripción GATA3/metabolismo , Ratones , Regiones Promotoras Genéticas , Activación Transcripcional
9.
Development ; 142(15): 2696-703, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26243870

RESUMEN

Despite the high occurrence of congenital abnormalities of the lower urinary tract in humans, the molecular, cellular and morphological aspects of their development are still poorly understood. Here, we use a conditional knockout approach to inactivate within the nephric duct (ND) lineage the two effectors of the Hippo pathway, Yap and Taz. Deletion of Yap leads to hydronephrotic kidneys with blind-ending megaureters at birth. In Yap mutants, the ND successfully migrates towards, and contacts, the cloaca. However, close analysis reveals that the tip of the Yap(-/-) ND forms an aberrant connection with the cloaca and does not properly insert into the cloaca, leading to later detachment of the ND from the cloaca. Taz deletion from the ND does not cause any defect, but analysis of Yap(-/-);Taz(-/-) NDs indicates that both genes play partially redundant roles in ureterovesical junction formation. Aspects of the Yap(-/-) phenotype resemble hypersensitivity to RET signaling, including excess budding of the ND, increased phospho-ERK and increased expression of Crlf1, Sprouty1, Etv4 and Etv5. Importantly, the Yap(ND) (-/-) ND phenotype can be largely rescued by reducing Ret gene dosage. Taken together, these results suggest that disrupting Yap/Taz activities enhances Ret pathway activity and contributes to pathogenesis of lower urinary tract defects in human infants.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Morfogénesis/fisiología , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/fisiología , Sistema Urinario/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Bromodesoxiuridina , Proteínas de Ciclo Celular , Técnica del Anticuerpo Fluorescente , Galactósidos , Técnicas de Inactivación de Genes , Humanos , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Indoles , Ratones , Fosfoproteínas/genética , Transactivadores , Proteínas Señalizadoras YAP
10.
Semin Cell Dev Biol ; 44: 97-106, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26410163

RESUMEN

The execution of developmental programs entails specific spatio-temporal expression of transcriptional regulators that ultimately control tissue morphogenesis and embryo patterning. Pax transcription factors are sequence-specific DNA-binding proteins exerting such regulatory activity in several tissues. In the urogenital system, Pax2 and Pax8 have emerged as crucial players at multiple steps of kidney and urinary tract development. They are involved in important processes such as cell survival, cell lineage decisions and tissue interactions through the regulation of sophisticated gene regulatory networks. Pax2/8 have additionally been directly associated with Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) and renal cancers in human. In this review, we provide an overview of landmark contributions to the understanding of Pax gene function in urinary tract development and disease with an emphasis on recent advances in the field.


Asunto(s)
Riñón/fisiología , Factores de Transcripción Paired Box/fisiología , Animales , Humanos , Riñón/embriología , Riñón/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo
11.
Semin Cell Dev Biol ; 36: 13-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25220017

RESUMEN

The elaboration of functional kidneys during embryonic development proceeds in a stepwise manner, starting with the formation of the embryonic pro- and mesonephros, followed by the induction and growth of the final metanephric kidney. These early stages of urinary tract development are critical for the embryo as a failure in pro/mesonephros morphogenesis leads to major developmental defects, often incompatible with life. The formation of the pro/mesonephros and its central component the nephric duct, is also interesting as it offers a relatively simple system to study cell biological behaviours underlying tissue morphogenesis. This system is especially well adapted to study the questions of cell lineage specification, epithelial integrity and plasticity, tissue interactions, collective cell migration/guidance and programmed cell death. In this review, we establish the link between these cell behaviours, their molecular regulators and early genitourinary tract development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/embriología , Mesonefro/embriología , Organogénesis/fisiología , Uréter/embriología , Desarrollo Embrionario , Células Epiteliales/citología , Humanos , Anomalías Urogenitales , Reflujo Vesicoureteral
12.
Development ; 140(16): 3413-22, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863482

RESUMEN

Leukocyte antigen related (LAR) family receptor protein tyrosine phosphatases (RPTPs) regulate the fine balance between tyrosine phosphorylation and dephosphorylation that is crucial for cell signaling during development and tissue homeostasis. Here we show that LAR RPTPs are required for normal development of the mandibular and maxillary regions. Approximately half of the mouse embryos lacking both Ptprs (RPTPσ) and Ptprf (LAR) exhibit micrognathia (small lower jaw), cleft palate and microglossia/glossoptosis (small and deep tongue), a phenotype closely resembling Pierre-Robin sequence in humans. We show that jaw bone and cartilage patterning occurs aberrantly in LAR family phosphatase-deficient embryos and that the mandibular arch harbors a marked decrease in cell proliferation. Analysis of signal transduction in embryonic tissues and mouse embryonic fibroblast cultures identifies an increase in Bmp-Smad signaling and an abrogation of canonical Wnt signaling associated with loss of the LAR family phosphatases. A reactivation of ß-catenin signaling by chemical inhibition of GSK3ß successfully resensitizes LAR family phosphatase-deficient cells to Wnt induction, indicating that RPTPs are necessary for normal Wnt/ß-catenin pathway activation. Together these results identify LAR RPTPs as important regulators of craniofacial morphogenesis and provide insight into the etiology of Pierre-Robin sequence.


Asunto(s)
Silenciador del Gen , Síndrome de Pierre Robin/enzimología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Animales , Proteína Axina/genética , Proteína Axina/metabolismo , Tipificación del Cuerpo , Desarrollo Óseo , Proliferación Celular , Células Cultivadas , Fisura del Paladar/enzimología , Fisura del Paladar/patología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Fibroblastos/enzimología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Indoles/farmacología , Mesodermo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Micrognatismo/enzimología , Micrognatismo/patología , Oximas/farmacología , Síndrome de Pierre Robin/patología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Vía de Señalización Wnt
13.
Hum Mol Genet ; 22(12): 2400-10, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23428429

RESUMEN

Loss of the tumor suppressor PTEN is a common occurrence in prostate cancer. This aberration leads to the ectopic activation of the PI3K-Akt pathway, which promotes tumor growth. Here, we show that the transcription factor Gata3 is progressively lost in Pten-deficient mouse prostate tumors as a result of both transcriptional down-regulation and increased proteasomal degradation. To determine the significance of this loss, we used conditional loss- and gain-of-function approaches to manipulate Gata3 expression levels in prostate tumors. Our results show that Gata3 inactivation in Pten-deficient prostates accelerates tumor invasion. Conversely, enforced expression of GATA3 in Pten-deficient tissues markedly delays tumor progression. In Pten-deficient prostatic ducts, enforced GATA3 prevented Akt activation, which correlated with the down-regulation of Pik3cg and Pik3c2a mRNAs, encoding respectively class I and II PI3K subunits. Remarkably, the majority of human prostate tumors similarly show loss of active GATA3 as they progress to the aggressive castrate-resistant stage. In addition, GATA3 expression levels in hormone-sensitive tumors holds predictive value for tumor recurrence. Together, these data establish Gata3 as an important regulator of prostate cancer progression.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Fosfohidrolasa PTEN/deficiencia , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Regulación hacia Abajo , Femenino , Factor de Transcripción GATA3/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
14.
Dev Biol ; 382(2): 555-66, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23920117

RESUMEN

Translating the developmental program encoded in the genome into cellular and morphogenetic functions requires the deployment of elaborate gene regulatory networks (GRNs). GRNs are especially crucial at the onset of organ development where a few regulatory signals establish the different programs required for tissue organization. In the renal system primordium (the pro/mesonephros), important regulators have been identified but their hierarchical and regulatory organization is still elusive. Here, we have performed a detailed analysis of the GRN underlying mouse pro/mesonephros development. We find that a core regulatory subcircuit composed of Pax2/8, Gata3 and Lim1 turns on a deeper layer of transcriptional regulators while activating effector genes responsible for cell signaling and tissue organization. Among the genes directly affected by the core components are the key developmental molecules Nephronectin (Npnt) and Plac8. Hence, the pro/mesonephros GRN links together several essential genes regulating tissue morphogenesis. This renal GRN sheds new light on the disease group Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) in that gene mutations are expected to generate different phenotypic outcomes as a consequence of regulatory network deficiencies rather than threshold effects from single genes.


Asunto(s)
Factor de Transcripción GATA3/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas con Homeodominio LIM/genética , Mesonefro/embriología , Factor de Transcripción PAX2/genética , Factores de Transcripción Paired Box/genética , Factores de Transcripción/genética , Animales , Línea Celular , Riñón/anomalías , Mesonefro/metabolismo , Ratones , Morfogénesis/genética , Factor de Transcripción PAX8
15.
Development ; 138(10): 2089-97, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21521737

RESUMEN

Urinary tract development depends on a complex series of events in which the ureter moves from its initial branch point on the nephric duct (ND) to its final insertion site in the cloaca (the primitive bladder and urethra). Defects in this maturation process can result in malpositioned ureters and hydronephrosis, a common cause of renal disease in children. Here, we report that insertion of the ND into the cloaca is an unrecognized but crucial step that is required for proper positioning of the ureter and that depends on Ret signaling. Analysis of Ret mutant mice at birth reveals hydronephrosis and defective ureter maturation, abnormalities that our results suggest are caused, at least in part, by delayed insertion of the ND. We find a similar set of malformations in mutants lacking either Gata3 or Raldh2. We show that these factors act in parallel to regulate ND insertion via Ret. Morphological analysis of ND extension in wild-type embryos reveals elaborate cellular protrusions at ND tips that are not detected in Ret, Gata3 or Raldh2 mutant embryos, suggesting that these protrusions may normally be important for fusion with the cloaca. Together, our studies reveal a novel Ret-dependent event, ND insertion, that, when abnormal, can cause obstruction and hydronephrosis at birth; whether ND defects underlie similar types of urinary tract abnormalities in humans is an interesting possibility.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Factor de Transcripción GATA3/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Sistema Urinario/embriología , Sistema Urinario/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Secuencia de Bases , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Cloaca/anomalías , Cloaca/embriología , Cloaca/metabolismo , Cartilla de ADN/genética , Femenino , Factor de Transcripción GATA3/deficiencia , Factor de Transcripción GATA3/genética , Regulación del Desarrollo de la Expresión Génica , Hidronefrosis/embriología , Hidronefrosis/genética , Hidronefrosis/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Embarazo , Proteínas Proto-Oncogénicas c-ret/deficiencia , Proteínas Proto-Oncogénicas c-ret/genética , Transducción de Señal , Sistema Urinario/anomalías
16.
J Immunol ; 188(9): 4535-42, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22450807

RESUMEN

Acute lung injury during bacterial infection is associated with neutrophilic inflammation, epithelial cell apoptosis, and disruption of the alveolar-capillary barrier. TLR4 is required for lung injury in animals exposed to bacterial LPS and initiates proinflammatory responses in part via the transcription factor NF-κB. Ligation of TLR4 also initiates a proapoptotic response by activating IFN-ß and STAT1-dependent genes. We recently demonstrated that mammalian target of rapamycin (mTOR), a key controller of cell growth and survival, can physically interact with STAT1 and suppress the induction of STAT1-dependent apoptosis genes. We therefore hypothesized that the mTOR inhibitor rapamycin would increase LPS-induced apoptosis and lung injury in vivo. Rapamycin increased lung injury and cellular apoptosis in C57BL/6J mice exposed to intratracheal LPS for 24 h. Rapamycin also augmented STAT1 activation, and the induction of STAT1-dependent genes that mediate cellular apoptosis (i.e., Fas, caspase-3). LPS-induced lung injury was attenuated in STAT1 knockout mice. In addition, LPS and IFN-ß-induced apoptosis was absent in cultured cells lacking STAT1, and, unlike in wild-type cells, a permissive effect of rapamycin was not observed. In contrast to its effect on STAT1, rapamycin inhibited NF-κB activation in vivo and reduced selected markers of inflammation (i.e., neutrophils in the bronchoalveolar lavage fluid, TNF-α). Therefore, although it inhibits NF-κB and neutrophilic inflammation, rapamycin augments LPS-induced lung injury and apoptosis in a mechanism that involves STAT1 and the induction of STAT1-dependent apoptosis genes.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Apoptosis/efectos de los fármacos , Lipopolisacáridos/toxicidad , Serina-Treonina Quinasas TOR/inmunología , Receptor Toll-Like 4/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Antibacterianos/farmacología , Apoptosis/genética , Apoptosis/inmunología , Lavado Broncoalveolar , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interferón beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
17.
Pediatr Nephrol ; 29(4): 637-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24104595

RESUMEN

Animal development progresses through the stepwise deployment of gene regulatory networks (GRN) encoded in the genome. Comparative analyses in different species and organ systems have revealed that GRN blueprints are composed of subcircuits with stereotypical architectures that are often reused as modular units. In this review, we report the evidence for the GRN underlying renal primordium development. In vertebrates, renal development is initiated by the induction of a field of intermediate mesoderm cells competent to undergo lineage specification and nephric (Wolffian) duct formation. Definition of the renal field leads to the activation of a core regulatory subcircuit composed of the transcription factors Pax2/8, Gata3 and Lim1. These transcription factors turn on a second layer of transcriptional regulators while also activating effectors of tissue morphogenesis and cellular specialization. Elongation and connection of the nephric duct to the cloaca (bladder/urethra primordium) is followed by metanephric kidney induction through signals emanating from the metanephric mesenchyme. Central to this process is the activation and positioning of the glial cell line-derived neurotrophic factor (Gdnf)-Ret signaling pathway by network subcircuits located in the mesenchyme and epithelial tissues of the caudal trunk. Evidence shows that each step of the renal primordium developmental program is regulated by structured GRN subunits organized in a hierarchical manner. Understanding the structure and dynamics of the renal GRN will help us understand the intrinsic phenotypical variability of congenital anomalies of the kidney and urinary tract and guide our approaches to regenerative medicine.


Asunto(s)
Desarrollo Embrionario/genética , Redes Reguladoras de Genes/fisiología , Riñón/embriología , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos
18.
Mol Biol Cell ; 35(2): br5, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37991903

RESUMEN

Loss of cell polarity and disruption of tissue organization are key features of tumorigenesis that are intrinsically linked to spindle orientation. Epithelial tumors are often characterized by spindle orientation defects, but how these defects impact tumor formation driven by common oncogenic mutations is not fully understood. Here, we examine the role of spindle orientation in adult epidermis by deleting a key spindle regulator, LGN, in normal tissue and in a PTEN-deficient mouse model. We report that LGN deficiency in PTEN mutant epidermis leads to a threefold increase in the likelihood of developing tumors on the snout, and an over 10-fold increase in tumor burden. In this tissue, loss of LGN alone increases perpendicular and oblique divisions of epidermal basal cells, at the expense of a planar orientation of division. PTEN loss alone does not significantly affect spindle orientation in these cells, but the combined loss of PTEN and LGN fully randomizes basal spindle orientation. A subset of LGN- and PTEN-deficient animals have increased amounts of proliferative spinous cells, which may be associated with tumorigenesis. These results indicate that loss of LGN impacts spindle orientation and accelerates epidermal tumorigenesis in a PTEN-deficient mouse model.


Asunto(s)
Epidermis , Huso Acromático , Animales , Ratones , Huso Acromático/genética , Células Epidérmicas , Carcinogénesis , Polaridad Celular/genética
19.
J Am Soc Nephrol ; 23(1): 63-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22021707

RESUMEN

ß-Catenin/Wnt signaling is essential during early inductive stages of kidney development, but its role during postinductive stages of nephron development and maturation is not well understood. In this study, we used Pax8Cre mice to target ß-catenin deficiency to renal epithelial cells at the late S-shaped body stage and the developing collecting ducts. The conditional ß-catenin knockout mice formed abnormal kidneys and had reduced renal function. The kidneys were hypoplastic with a thin cortex; a superficial layer of tubules was missing. A high proportion of glomeruli had small, underdeveloped capillary tufts. In these glomeruli, well differentiated podocytes replaced parietal epithelial cells in Bowman's capsule; capillaries toward the outer aspect of these podocytes mimicked the formation of glomerular capillaries. Tracing nephrogenesis in embryonic conditional ß-catenin knockout mice revealed that these "parietal podocytes" derived from precursor cells in the parietal layer of the S-shaped body by direct lineage switch. Taken together, these findings demonstrate that ß-catenin/Wnt signaling is important during the late stages of nephrogenesis and for the lineage specification of parietal epithelial cells.


Asunto(s)
Riñón/embriología , Organogénesis , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Epiteliales/citología , Riñón/citología , Riñón/metabolismo , Ratones , Ratones Noqueados , beta Catenina/genética
20.
Cell Rep ; 42(3): 112202, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36871220

RESUMEN

In developing embryos, specific cell populations are often removed to remodel tissue architecture for organogenesis. During urinary tract development, an epithelial duct called the common nephric duct (CND) gets shortened and eventually eliminated to remodel the entry point of the ureter into the bladder. Here we show that non-professional efferocytosis (the process in which epithelial cells engulf apoptotic bodies) is the main mechanism that contributes to CND shortening. Combining biological metrics and computational modeling, we show that efferocytosis with actomyosin contractility are essential factors that drive the CND shortening without compromising the ureter-bladder structural connection. The disruption of either apoptosis, non-professional efferocytosis, or actomyosin results in contractile tension reduction and deficient CND shortening. Actomyosin activity helps to maintain tissue architecture while non-professional efferocytosis removes cellular volume. Together our results demonstrate that non-professional efferocytosis with actomyosin contractility are important morphogenetic factors controlling CND morphogenesis.


Asunto(s)
Actomiosina , Células Epiteliales , Fagocitosis , Epitelio , Morfogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA