Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Development ; 149(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35905011

RESUMEN

Smooth muscle cells (SMCs) are a crucial component of the mesenchymal wall of the ureter, as they account for the efficient removal of the urine from the renal pelvis to the bladder by means of their contractile activity. Here, we show that the zinc-finger transcription factor gene Gata6 is expressed in mesenchymal precursors of ureteric SMCs under the control of BMP4 signaling. Mice with a conditional loss of Gata6 in these precursors exhibit a delayed onset and reduced level of SMC differentiation and peristaltic activity, as well as dilatation of the ureter and renal pelvis (hydroureternephrosis) at birth and at postnatal stages. Molecular profiling revealed a delayed and reduced expression of the myogenic driver gene Myocd, but the activation of signaling pathways and transcription factors previously implicated in activation of the visceral SMC program in the ureter was unchanged. Additional gain-of-function experiments suggest that GATA6 cooperates with FOXF1 in Myocd activation and SMC differentiation, possibly as pioneer and lineage-determining factors, respectively.


Asunto(s)
Uréter , Animales , Diferenciación Celular/genética , Ratones , Desarrollo de Músculos , Músculo Liso , Miocitos del Músculo Liso/fisiología , Uréter/metabolismo
2.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35103284

RESUMEN

The contractile phenotype of smooth muscle cells (SMCs) is transcriptionally controlled by a complex of the DNA-binding protein SRF and the transcriptional co-activator MYOCD. The pathways that activate expression of Myocd and of SMC structural genes in mesenchymal progenitors are diverse, reflecting different intrinsic and extrinsic signaling inputs. Taking the ureter as a model, we analyzed whether Notch signaling, a pathway previously implicated in vascular SMC development, also affects visceral SMC differentiation. We show that mice with a conditional deletion of the unique Notch mediator RBPJ in the undifferentiated ureteric mesenchyme exhibit altered ureter peristalsis with a delayed onset, and decreased contraction frequency and intensity at fetal stages. They also develop hydroureter 2 weeks after birth. Notch signaling is required for precise temporal activation of Myocd expression and, independently, for expression of a group of late SMC structural genes. Based on additional expression analyses, we suggest that a mesenchymal JAG1-NOTCH2/NOTCH3 module regulates visceral SMC differentiation in the ureter in a biphasic and bimodal manner, and that its molecular function differs from that in the vascular system.


Asunto(s)
Diferenciación Celular , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Uréter/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diaminas/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Tiazoles/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Uréter/citología , Uréter/crecimiento & desarrollo , Vísceras/citología , Vísceras/metabolismo
3.
Development ; 148(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33795231

RESUMEN

All epithelial components of the inner ear, including sensory hair cells and innervating afferent neurons, arise by patterning and differentiation of epithelial progenitors residing in a simple sphere, the otocyst. Here, we identify the transcriptional repressors TBX2 and TBX3 as novel regulators of these processes in the mouse. Ablation of Tbx2 from the otocyst led to cochlear hypoplasia, whereas loss of Tbx3 was associated with vestibular malformations. The loss of function of both genes (Tbx2/3cDKO) prevented inner ear morphogenesis at midgestation, resulting in indiscernible cochlear and vestibular structures at birth. Morphogenetic impairment occurred concomitantly with increased apoptosis in ventral and lateral regions of Tbx2/3cDKO otocysts around E10.5. Expression analyses revealed partly disturbed regionalisation, and a posterior-ventral expansion of the neurogenic domain in Tbx2/3cDKO otocysts at this stage. We provide evidence that repression of FGF signalling by TBX2 is important to restrict neurogenesis to the anterior-ventral otocyst and implicate another T-box factor, TBX1, as a crucial mediator in this regulatory network.


Asunto(s)
Apoptosis , Oído Interno/embriología , Regulación del Desarrollo de la Expresión Génica , Organogénesis , Transducción de Señal , Proteínas de Dominio T Box/biosíntesis , Animales , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/genética
4.
Biochem J ; 479(1): 91-109, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-34935912

RESUMEN

The TBX18 transcription factor regulates patterning and differentiation programs in the primordia of many organs yet the molecular complexes in which TBX18 resides to exert its crucial transcriptional function in these embryonic contexts have remained elusive. Here, we used 293 and A549 cells as an accessible cell source to search for endogenous protein interaction partners of TBX18 by an unbiased proteomic approach. We tagged endogenous TBX18 by CRISPR/Cas9 targeted genome editing with a triple FLAG peptide, and identified by anti-FLAG affinity purification and subsequent LC-MS analysis the ZMYM2 protein to be statistically enriched together with TBX18 in both 293 and A549 nuclear extracts. Using a variety of assays, we confirmed the binding of TBX18 to ZMYM2, a component of the CoREST transcriptional corepressor complex. Tbx18 is coexpressed with Zmym2 in the mesenchymal compartment of the developing ureter of the mouse, and mutations in TBX18 and in ZMYM2 were recently linked to congenital anomalies in the kidney and urinary tract (CAKUT) in line with a possible in vivo relevance of TBX18-ZMYM2 protein interaction in ureter development.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteómica/métodos , Transducción de Señal/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Células A549 , Animales , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Ratones , Mutación , Embarazo , Unión Proteica , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Transfección , Uréter/embriología , Uréter/metabolismo , Anomalías Urogenitales/genética , Anomalías Urogenitales/metabolismo , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/metabolismo
5.
Hum Mol Genet ; 29(18): 3064-3080, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-32886109

RESUMEN

ANKS6 is a ciliary protein that localizes to the proximal compartment of the primary cilium, where it regulates signaling. Mutations in the ANKS6 gene cause multiorgan ciliopathies in humans, which include laterality defects of the visceral organs, renal cysts as part of nephronophthisis and congenital hepatic fibrosis (CHF) in the liver. Although CHF together with liver ductal plate malformations are common features of several human ciliopathy syndromes, including nephronophthisis-related ciliopathies, the mechanism by which mutations in ciliary genes lead to bile duct developmental abnormalities is not understood. Here, we generated a knockout mouse model of Anks6 and show that ANKS6 function is required for bile duct morphogenesis and cholangiocyte differentiation. The loss of Anks6 causes ciliary abnormalities, ductal plate remodeling defects and periportal fibrosis in the liver. Our expression studies and biochemical analyses show that biliary abnormalities in Anks6-deficient livers result from the dysregulation of YAP transcriptional activity in the bile duct-lining epithelial cells. Mechanistically, our studies suggest, that ANKS6 antagonizes Hippo signaling in the liver during bile duct development by binding to Hippo pathway effector proteins YAP1, TAZ and TEAD4 and promoting their transcriptional activity. Together, this study reveals a novel function for ANKS6 in regulating Hippo signaling during organogenesis and provides mechanistic insights into the regulatory network controlling bile duct differentiation and morphogenesis during liver development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Hígado/crecimiento & desarrollo , Proteínas Musculares/genética , Factores de Transcripción/genética , Animales , Conductos Biliares/crecimiento & desarrollo , Conductos Biliares/metabolismo , Conductos Biliares/patología , Diferenciación Celular/genética , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Humanos , Hígado/anomalías , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Morfogénesis/genética , Transducción de Señal/genética , Factores de Transcripción de Dominio TEA , Proteínas Señalizadoras YAP
6.
Development ; 145(23)2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30478225

RESUMEN

The organized array of smooth muscle cells (SMCs) and fibroblasts in the walls of visceral tubular organs arises by patterning and differentiation of mesenchymal progenitors surrounding the epithelial lumen. Here, we show that the TBX2 and TBX3 transcription factors have novel and required roles in regulating these processes in the murine ureter. Co-expression of TBX2 and TBX3 in the inner mesenchymal region of the developing ureter requires canonical WNT signaling. Loss of TBX2/TBX3 in this region disrupts activity of two crucial drivers of the SMC program, Foxf1 and BMP4 signaling, resulting in decreased SMC differentiation and increased extracellular matrix. Transcriptional profiling and chromatin immunoprecipitation experiments revealed that TBX2/TBX3 directly repress expression of the WNT antagonists Dkk2 and Shisa2, the BMP antagonist Bmper and the chemokine Cxcl12 These findings suggest that TBX2/TBX3 are effectors of canonical WNT signaling in the ureteric mesenchyme that promote SMC differentiation by maintaining BMP4 and WNT signaling in the inner region, while restricting CXCL12 signaling to the outer layer of fibroblast-fated mesenchyme.


Asunto(s)
Tipificación del Cuerpo , Diferenciación Celular , Mesodermo/embriología , Proteínas de Dominio T Box/metabolismo , Uréter/embriología , Vía de Señalización Wnt , Animales , Proteína Morfogenética Ósea 4/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Ratones , Modelos Biológicos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Peristaltismo , Proteínas de Dominio T Box/genética , Transcriptoma/genética , Uréter/metabolismo , Uréter/patología
7.
Respir Res ; 22(1): 85, 2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731112

RESUMEN

BACKGROUND: Tbx2 encodes a transcriptional repressor implicated in the development of numerous organs in mouse. During lung development TBX2 maintains the proliferation of mesenchymal progenitors, and hence, epithelial proliferation and branching morphogenesis. The pro-proliferative function was traced to direct repression of the cell-cycle inhibitor genes Cdkn1a and Cdkn1b, as well as of genes encoding WNT antagonists, Frzb and Shisa3, to increase pro-proliferative WNT signaling. Despite these important molecular insights, we still lack knowledge of the DNA occupancy of TBX2 in the genome, and of the protein interaction partners involved in transcriptional repression of target genes. METHODS: We used chromatin immunoprecipitation (ChIP)-sequencing and expression analyses to identify genomic DNA-binding sites and transcription units directly regulated by TBX2 in the developing lung. Moreover, we purified TBX2 containing protein complexes from embryonic lung tissue and identified potential interaction partners by subsequent liquid chromatography/mass spectrometry. The interaction with candidate proteins was validated by immunofluorescence, proximity ligation and individual co-immunoprecipitation analyses. RESULTS: We identified Il33 and Ccn4 as additional direct target genes of TBX2 in the pulmonary mesenchyme. Analyzing TBX2 occupancy data unveiled the enrichment of five consensus sequences, three of which match T-box binding elements. The remaining two correspond to a high mobility group (HMG)-box and a homeobox consensus sequence motif. We found and validated binding of TBX2 to the HMG-box transcription factor HMGB2 and the homeobox transcription factor PBX1, to the heterochromatin protein CBX3, and to various members of the nucleosome remodeling and deacetylase (NuRD) chromatin remodeling complex including HDAC1, HDAC2 and CHD4. CONCLUSION: Our data suggest that TBX2 interacts with homeobox and HMG-box transcription factors as well as with the NuRD chromatin remodeling complex to repress transcription of anti-proliferative genes in the pulmonary mesenchyme.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genómica , Pulmón/metabolismo , Proteómica , Proteínas de Dominio T Box/metabolismo , Animales , Sitios de Unión , Proteínas CCN de Señalización Intercelular/genética , Proteínas CCN de Señalización Intercelular/metabolismo , Proliferación Celular , Secuenciación de Inmunoprecipitación de Cromatina , Cromatografía Liquida , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células HEK293 , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Humanos , Interleucina-33/genética , Interleucina-33/metabolismo , Pulmón/embriología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Proteínas de Dominio T Box/genética , Espectrometría de Masas en Tándem
8.
J Pathol ; 248(4): 452-463, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30916783

RESUMEN

The establishment of the peristaltic machinery of the ureter is precisely controlled to cope with the onset of urine production in the fetal kidney. Retinoic acid (RA) has been identified as a signal that maintains the mesenchymal progenitors of the contractile smooth muscle cells (SMCs), while WNTs, SHH, and BMP4 induce their differentiation. How the activity of the underlying signalling pathways is controlled in time, space, and quantity to activate coordinately the SMC programme is poorly understood. Here, we provide evidence that the Zn-finger transcription factor GATA2 is involved in this crosstalk. In mice, Gata2 is expressed in the undifferentiated ureteric mesenchyme under control of RA signalling. Conditional deletion of Gata2 by a Tbx18cre driver results in hydroureter formation at birth, associated with a loss of differentiated SMCs. Analysis at earlier stages and in explant cultures revealed that SMC differentiation is not abrogated but delayed and that dilated ureters can partially regain peristaltic activity when relieved of urine pressure. Molecular analysis identified increased RA signalling as one factor contributing to the delay in SMC differentiation, possibly caused by reduced direct transcriptional activation of Cyp26a1, which encodes an RA-degrading enzyme. Our study identified GATA2 as a feedback inhibitor of RA signalling important for precise onset of ureteric SMC differentiation, and suggests that in a subset of cases of human congenital ureter dilatations, temporary relief of urine pressure may ameliorate the differentiation status of the SMC coat. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Diferenciación Celular , Factor de Transcripción GATA2/deficiencia , Mesodermo/embriología , Miocitos del Músculo Liso/fisiología , Uréter/embriología , Enfermedades Ureterales/embriología , Animales , Biomarcadores/metabolismo , Femenino , Factor de Transcripción GATA2/genética , Masculino , Mesodermo/metabolismo , Ratones , Transducción de Señal , Tretinoina/metabolismo , Uréter/anomalías , Uréter/metabolismo , Enfermedades Ureterales/congénito , Enfermedades Ureterales/metabolismo
9.
PLoS Genet ; 13(8): e1006951, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28797033

RESUMEN

The differentiated cell types of the epithelial and mesenchymal tissue compartments of the mature ureter of the mouse arise in a precise temporal and spatial sequence from uncommitted precursor cells of the distal ureteric bud epithelium and its surrounding mesenchyme. Previous genetic efforts identified a member of the Hedgehog (HH) family of secreted proteins, Sonic hedgehog (SHH) as a crucial epithelial signal for growth and differentiation of the ureteric mesenchyme. Here, we used conditional loss- and gain-of-function experiments of the unique HH signal transducer Smoothened (SMO) to further characterize the cellular functions and unravel the effector genes of HH signaling in ureter development. We showed that HH signaling is not only required for proliferation and SMC differentiation of cells of the inner mesenchymal region but also for survival of cells of the outer mesenchymal region, and for epithelial proliferation and differentiation. We identified the Forkhead transcription factor gene Foxf1 as a target of HH signaling in the ureteric mesenchyme. Expression of a repressor version of FOXF1 in this tissue completely recapitulated the mesenchymal and epithelial proliferation and differentiation defects associated with loss of HH signaling while re-expression of a wildtype version of FOXF1 in the inner mesenchymal layer restored these cellular programs when HH signaling was inhibited. We further showed that expression of Bmp4 in the ureteric mesenchyme depends on HH signaling and Foxf1, and that exogenous BMP4 rescued cell proliferation and epithelial differentiation in ureters with abrogated HH signaling or FOXF1 function. We conclude that SHH uses a FOXF1-BMP4 module to coordinate the cellular programs for ureter elongation and differentiation, and suggest that deregulation of this signaling axis occurs in human congenital anomalies of the kidney and urinary tract (CAKUT).


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Uréter/embriología , Animales , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Epitelio/embriología , Epitelio/metabolismo , Femenino , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/genética , Procesamiento de Imagen Asistido por Computador , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Análisis por Micromatrices , Organogénesis/genética , Reproducibilidad de los Resultados , Transducción de Señal , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Uréter/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L767-L783, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30702346

RESUMEN

The mesothelial lining of the lung, the visceral pleura, and of the heart, the epicardium, derive from a common multipotent precursor tissue, the mesothelium of the embryonic thoracic cavity that also contributes to organ-specific mesenchymal cell types. Insight into mesothelial mobilization and differentiation has prevailedin the developing heart while the mesenchymal transition and fate of the visceral pleura are poorly understood. Here, we use the fact that the early mesothelium of both the lung and the heart expresses the transcription factor gene Wt1, to comparatively analyze mesothelial mobilization in the two organs by a genetic cre-loxP-based conditional approach. We show that epicardial cells are mobilized in a large number between E12.5 and E14.5, whereas pleural mobilization occurs only sporadically and variably in few regions of the lung in a temporally highly confined manner shortly after E12.5. Mesothelium-specific inactivation of unique pathway components using a Wt1creERT2 line excluded a requirement for canonical WNT, NOTCH, HH, TGFB, PDGFRA, and FGFR1/FGFR2 signaling in the mesenchymal transition of the visceral pleura but indicated a deleterious effect of activated WNT, NOTCH, and HH signaling on lung development. Epicardial mobilization was negatively impacted on by loss of HH, PDGFRA, FGFR1/2 signaling. Epicardial overactivation of WNT, NOTCH, and HH disturbed epicardial and myocardial integrity. We conclude that mesothelial mobilization in the developing lung and heart differs in timing, quantity and pathway dependency, indicating the organ specificity of the program.


Asunto(s)
Epitelio/embriología , Corazón/embriología , Pulmón/embriología , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Epitelio/metabolismo , Femenino , Edad Gestacional , Inmunohistoquímica , Pulmón/metabolismo , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Miocardio/metabolismo , Embarazo , Transducción de Señal/genética , Proteínas WT1/deficiencia , Proteínas WT1/genética , Proteínas WT1/metabolismo , Vía de Señalización Wnt/genética
11.
Respir Res ; 20(1): 292, 2019 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-31870435

RESUMEN

BACKGROUND: In the embryonic mammalian lung, mesenchymal cells act both as a signaling center for epithelial proliferation, differentiation and morphogenesis as well as a source for a multitude of differentiated cell types that support the structure of the developing and mature organ. Whether the embryonic pulmonary mesenchyme is a homogenous precursor pool and how it diversifies into different cell lineages is poorly understood. We have previously shown that the T-box transcription factor gene Tbx2 is expressed in the pulmonary mesenchyme of the developing murine lung and is required therein to maintain branching morphogenesis. METHODS: We determined Tbx2/TBX2 expression in the developing murine lung by in situ hybridization and immunofluorescence analyses. We used a genetic lineage tracing approach with a Cre line under the control of endogenous Tbx2 control elements (Tbx2cre), and the R26mTmG reporter line to trace TBX2-positive cells in the murine lung. We determined the fate of the TBX2 lineage by co-immunofluorescence analysis of the GFP reporter and differentiation markers in normal murine lungs and in lungs lacking or overexpressing TBX2 in the pulmonary mesenchyme. RESULTS: We show that TBX2 is strongly expressed in mesenchymal progenitors in the developing murine lung. In differentiated smooth muscle cells and in fibroblasts, expression of TBX2 is still widespread but strongly reduced. In mesothelial and endothelial cells expression is more variable and scattered. All fetal smooth muscle cells, endothelial cells and fibroblasts derive from TBX2+ progenitors, whereas half of the mesothelial cells have a different descent. The fate of TBX2-expressing cells is not changed in Tbx2-deficient and in TBX2-constitutively overexpressing mice but the distribution and abundance of endothelial and smooth muscle cells is changed in the overexpression condition. CONCLUSION: The fate of pulmonary mesenchymal progenitors is largely independent of TBX2. Nevertheless, a successive and precisely timed downregulation of TBX2 is necessary to allow proper differentiation and functionality of bronchial smooth muscle cells and to limit endothelial differentiation. Our work suggests expression of TBX2 in an early pulmonary mesenchymal progenitor and supports a role of TBX2 in maintaining the precursor state of these cells.


Asunto(s)
Pulmón/embriología , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas de Dominio T Box/biosíntesis , Animales , Linaje de la Célula/fisiología , Células Cultivadas , Femenino , Pulmón/citología , Ratones , Ratones Transgénicos , Embarazo , Proteínas de Dominio T Box/genética
12.
PLoS Genet ; 9(1): e1003189, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23341776

RESUMEN

Vertebrate organ development relies on the precise spatiotemporal orchestration of proliferation rates and differentiation patterns in adjacent tissue compartments. The underlying integration of patterning and cell cycle control during organogenesis is insufficiently understood. Here, we have investigated the function of the patterning T-box transcription factor gene Tbx2 in lung development. We show that lungs of Tbx2-deficient mice are markedly hypoplastic and exhibit reduced branching morphogenesis. Mesenchymal proliferation was severely decreased, while mesenchymal differentiation into fibrocytes was prematurely induced. In the epithelial compartment, proliferation was reduced and differentiation of alveolar epithelial cells type 1 was compromised. Prior to the observed cellular changes, canonical Wnt signaling was downregulated, and Cdkn1a (p21) and Cdkn1b (p27) (two members of the Cip/Kip family of cell cycle inhibitors) were strongly induced in the Tbx2-deficient lung mesenchyme. Deletion of both Cdkn1a and Cdkn1b rescued, to a large degree, the growth deficits of Tbx2-deficient lungs. Prolongation of Tbx2 expression into adulthood led to hyperproliferation and maintenance of mesenchymal progenitor cells, with branching morphogenesis remaining unaffected. Expression of Cdkn1a and Cdkn1b was ablated from the lung mesenchyme in this gain-of-function setting. We further show by ChIP experiments that Tbx2 directly binds to Cdkn1a and Cdkn1b loci in vivo, defining these two genes as direct targets of Tbx2 repressive activity in the lung mesenchyme. We conclude that Tbx2-mediated regulation of Cdkn1a and Cdkn1b represents a crucial node in the network integrating patterning information and cell cycle regulation that underlies growth, differentiation, and branching morphogenesis of this organ.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Pulmón , Proteínas de Dominio T Box , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Mesodermo , Ratones , Morfogénesis , Transducción de Señal , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
13.
PLoS Genet ; 9(4): e1003467, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23633963

RESUMEN

Vertebrate limb outgrowth is driven by a positive feedback loop that involves Sonic hedgehog (Shh) and Gremlin1 (Grem1) in the posterior limb bud mesenchyme and Fibroblast growth factors (Fgfs) in the overlying epithelium. Proper spatio-temporal control of these signaling activities is required to avoid limb malformations such as polydactyly. Here we show that, in Tbx2-deficient hindlimbs, Shh/Fgf4 signaling is prolonged, resulting in increased limb bud size and duplication of digit 4. In turn, limb-specific Tbx2 overexpression leads to premature termination of this signaling loop with smaller limbs and reduced digit number as phenotypic manifestation. We show that Tbx2 directly represses Grem1 in distal regions of the posterior limb mesenchyme allowing Bone morphogenetic protein (Bmp) signaling to abrogate Fgf4/9/17 expression in the overlying epithelium. Since Tbx2 itself is a target of Bmp signaling, our data identify a growth-inhibiting positive feedback loop (Bmp/Tbx2/Grem1). We propose that proliferative expansion of Tbx2-expressing cells mediates self-termination of limb bud outgrowth due to their refractoriness to Grem1 induction.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Proteínas Hedgehog/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Esbozos de los Miembros/crecimiento & desarrollo , Proteínas de Dominio T Box/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Citocinas , Epitelio/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Esbozos de los Miembros/metabolismo , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Fenotipo , Transducción de Señal , Proteínas de Dominio T Box/metabolismo
14.
J Hepatol ; 62(1): 101-10, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25135862

RESUMEN

BACKGROUND & AIMS: Current hepatic differentiation protocols for human embryonic stem cells (ESCs) require substantial improvements. MicroRNAs (miRNAs) have been reported to regulate hepatocyte cell fate during liver development, but their utility to improve hepatocyte differentiation from ESCs remains to be investigated. Therefore, our aim was to identify and to analyse hepatogenic miRNAs for their potential to improve hepatocyte differentiation from ESCs. METHODS: By miRNA profiling and in vitro screening, we identified miR-199a-5p among several potential hepatogenic miRNAs. Transplantation studies of miR-199a-5p-inhibited hepatocyte-like cells (HLCs) in the liver of immunodeficient fumarylacetoacetate hydrolase knockout mice (Fah(-/-)/Rag2(-/-)/Il2rg(-/-)) were performed to assess their in vivo liver repopulation potential. For target determination, western blot and luciferase reporter assay were carried out. RESULTS: miRNA profiling revealed 20 conserved candidate hepatogenic miRNAs. By miRNA screening, only miR-199a-5p inhibition in HLCs was found to be able to enhance the in vitro hepatic differentiation of mouse as well as human ESCs. miR-199a-5p inhibition in human ESCs-derived HLCs enhanced their engraftment and repopulation capacity in the liver of Fah(-/-)/Rag2(-/-)/Il2rg(-/-) mice. Furthermore, we identified SMARCA4 and MST1 as novel targets of miR-199a-5p that may contribute to the improved hepatocyte generation and in vivo liver repopulation. CONCLUSIONS: Our findings demonstrate that miR-199a-5p inhibition in ES-derived HLCs leads to improved hepatocyte differentiation. Upon transplantation, HLCs were able to engraft and repopulate the liver of Fah(-/-)/Rag2(-/-)/Il2rg(-/-) mice. Thus, our findings suggest that miRNA modulation may serve as a promising approach to generate more mature HLCs from stem cell sources for the treatment of liver diseases.


Asunto(s)
Regulación de la Expresión Génica , Hepatocitos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Trasplante de Hígado , MicroARNs/genética , ARN/genética , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Hepatocitos/citología , Células Madre Embrionarias Humanas/citología , Humanos , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Circ Res ; 108(7): 813-23, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21252157

RESUMEN

RATIONALE: The embryonic epicardium plays a crucial role in the formation of the coronary vasculature and in myocardial development, yet the exact contribution of epicardium-derived cells (EPDCs) to the vascular and connective tissue of the heart, and the factors that regulate epicardial differentiation, are insufficiently understood. OBJECTIVE: To define the role of Notch signaling in murine epicardial development. METHODS AND RESULTS: Using in situ hybridization and RT-PCR analyses, we detected expression of a number of Notch receptor and ligand genes in early epicardial development, as well as during formation of coronary arteries. Mice with epicardial deletion of Rbpj, the unique intracellular mediator of Notch signaling, survived to adulthood and exhibited enlarged coronary venous and arterial beds. Using a Tbx18-based genetic lineage tracing system, we show that EPDCs give rise to fibroblasts and coronary smooth muscle cells (SMCs) but not to endothelial cells in the wild type, whereas in Rbpj-deficient embryos EPDCs form and surround the developing arteries but fail to differentiate into SMCs. Conditional activation of Notch signaling results in premature SMC differentiation of epicardial cells and prevents coronary angiogenesis. We further show that Notch signaling regulates, and cooperates with transforming growth factor ß signaling in SM differentiation of EPDCs. CONCLUSIONS: Notch signaling is a crucial regulator of SM differentiation of EPDCs, and thus, of formation of a functional coronary system.


Asunto(s)
Diferenciación Celular/fisiología , Miocitos del Músculo Liso/citología , Pericardio/citología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Animales , Aterosclerosis/fisiopatología , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/fisiopatología , Femenino , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Pericardio/embriología , Pericardio/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores Notch/genética , Factor de Crecimiento Transformador beta/fisiología
16.
Nat Commun ; 13(1): 7628, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494345

RESUMEN

The auditory function of the mammalian cochlea relies on two types of mechanosensory hair cells and various non-sensory supporting cells. Recent studies identified the transcription factors INSM1 and IKZF2 as regulators of outer hair cell (OHC) fate. However, the transcriptional regulation of the differentiation of inner hair cells (IHCs) and their associated inner supporting cells (ISCs) has remained enigmatic. Here, we show that the expression of the transcription factor TBX2 is restricted to IHCs and ISCs from the onset of differentiation until adulthood and examine its function using conditional deletion and misexpression approaches in the mouse. We demonstrate that TBX2 acts in prosensory progenitors as a patterning factor by specifying the inner compartment of the sensory epithelium that subsequently gives rise to IHCs and ISCs. Hair cell-specific inactivation or misexpression causes transdifferentiation of hair cells indicating a cell-autonomous function of TBX2 in inducing and maintaining IHC fate.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas Internas , Ratones , Animales , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Cóclea/fisiología , Factores de Transcripción/metabolismo , Diferenciación Celular/genética , Órgano Espiral/metabolismo , Mamíferos/metabolismo
17.
Dev Biol ; 336(2): 145-55, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19769959

RESUMEN

Formation of the mammalian secondary palate is a highly regulated and complex process. Impairment of the underlying cellular and molecular programs often results in cleft palate, a common birth defect in mammals. Here we report that Tbx2 and Tbx3, two closely related genes encoding T-box transcription factors, are expressed in the mesenchyme of the mouse palatal structures during development. Mice homozygous mutant for Tbx2 and mice double heterozygous for Tbx2 and Tbx3 exhibit a cleft palate phenotype arguing for an important contribution of Tbx2 and Tbx3 to palatogenesis. In Tbx2-deficient embryos, the bilateral primordial palatal shelves form but are smaller and retarded in the outgrowth process. They do not make contact but retain the potential to fuse. Development of other craniofacial structures appears normal, suggesting that impaired palate formation in Tbx2-mutant mice is caused by a primary defect in the palatal shelf mesenchyme. This is further supported by increased cell proliferation and apoptosis accompanied by increased expression of Bmp4 and CyclinD1 in Tbx2-deficient palatal shelves. Hence, Tbx2 and Tbx3 function overlappingly to control growth of the palatal shelf mesenchyme.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hueso Paladar/embriología , Proteínas de Dominio T Box/genética , Animales , Proteína Morfogenética Ósea 4/genética , Ciclina D1/genética , Hibridación in Situ , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/fisiología
18.
Hepatology ; 49(3): 969-78, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19140222

RESUMEN

UNLABELLED: After specification of the hepatic endoderm, mammalian liver organogenesis progresses through a series of morphological stages that culminate in the migration of hepatocytes into the underlying mesenchyme to populate the hepatic lobes. Here, we show that in the mouse the transcriptional repressor Tbx3, a member of the T-box protein family, is required for the transition from a hepatic diverticulum with a pseudo-stratified epithelium to a cell-emergent liver bud. In Tbx3-deficient embryos, proliferation in the hepatic epithelium is severely reduced, hepatoblasts fail to delaminate, and cholangiocyte rather than hepatocyte differentiation occurs. Molecular analyses suggest that the primary function of Tbx3 is to maintain expression of hepatocyte transcription factors, including hepatic nuclear factor 4a (Hnf4a) and CCAAT/enhancer binding protein (C/EBP), alpha (Cebpa), and to repress expression of cholangiocyte transcription factors such as Onecut1 (Hnf6) and Hnf1b. CONCLUSION: Tbx3 controls liver bud expansion by suppressing cholangiocyte and favoring hepatocyte differentiation in the liver bud.


Asunto(s)
Conductos Biliares Extrahepáticos/embriología , Diferenciación Celular/fisiología , Células Epiteliales/citología , Hígado/embriología , Hígado/metabolismo , Organogénesis/fisiología , Proteínas de Dominio T Box/metabolismo , Animales , Conductos Biliares Extrahepáticos/citología , Conductos Biliares Extrahepáticos/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Epiteliales/metabolismo , Femenino , Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Hígado/citología , Ratones , Ratones Noqueados , Embarazo , Proteínas de Dominio T Box/genética
19.
Sci Rep ; 7(1): 14803, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093497

RESUMEN

The differentiated cell types of the mature ureter arise from the distal ureteric bud epithelium and its surrounding mesenchyme. Uncommitted epithelial cells first become intermediate cells from which both basal and superficial cells develop. Mesenchymal progenitors give rise to separated layers of adventitial fibrocytes, smooth muscle cells and lamina propria fibrocytes. How progenitor expansion and differentiation are balanced is poorly understood. Here, we addressed the role of retinoic acid (RA) signaling in these programs. Using expression analysis of components and target genes, we show that pathway activity is restricted to the mesenchymal and epithelial progenitor pools. Inhibition of RA signaling in ureter explant cultures resulted in tissue hypoplasia with a relative expansion of smooth muscle cells at the expense of lamina propria fibroblasts in the mesenchyme, and of superficial cells at the expense of intermediate cells in the ureteric epithelium. Administration of RA led to a slight reduction of smooth muscle cells, and almost completely prevented differentiation of intermediate cells into basal and superficial cells. We identified cellular programs and transcriptional targets of RA signaling that may account for this activity. We conclude that RA signaling is required and sufficient to maintain mesenchymal and epithelial progenitors in early ureter development.


Asunto(s)
Embrión de Mamíferos/embriología , Células Epiteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Uréter/embriología , Animales , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo
20.
Dev Cell ; 39(2): 239-253, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27720610

RESUMEN

Numerous signals drive the proliferative expansion of the distal endoderm and the underlying mesenchyme during lung branching morphogenesis, but little is known about how these signals are integrated. Here, we show by analysis of conditional double mutants that the two T-box transcription factor genes Tbx2 and Tbx3 act together in the lung mesenchyme to maintain branching morphogenesis. Expression of both genes depends on epithelially derived Shh signaling, with additional modulation by Bmp, Wnt, and Tgfß signaling. Genetic rescue experiments reveal that Tbx2 and Tbx3 function downstream of Shh to maintain pro-proliferative mesenchymal Wnt signaling, in part by direct repression of the Wnt antagonists Frzb and Shisa3. In combination with our previous finding that Tbx2 and Tbx3 repress the cell-cycle inhibitors Cdkn1a and Cdkn1b, we conclude that Tbx2 and Tbx3 maintain proliferation of the lung mesenchyme by way of at least two molecular mechanisms: regulating cell-cycle regulation and integrating the activity of multiple signaling pathways.


Asunto(s)
Proteínas Hedgehog/metabolismo , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Morfogénesis , Proteínas de Dominio T Box/metabolismo , Vía de Señalización Wnt , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Femenino , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA