Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Biol ; 18(10): e3000850, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33017398

RESUMEN

Cooperative DNA binding is a key feature of transcriptional regulation. Here we examined the role of cooperativity in Notch signaling by CRISPR-mediated engineering of mice in which neither Notch1 nor Notch2 can homo- or heterodimerize, essential for cooperative binding to sequence-paired sites (SPS) located near many Notch-regulated genes. Although most known Notch-dependent phenotypes were unaffected in Notch1/2 dimer-deficient mice, a subset of tissues proved highly sensitive to loss of cooperativity. These phenotypes include heart development, compromised viability in combination with low gene dose, and the gut, developing ulcerative colitis in response to 1% dextran sulfate sodium (DSS). The most striking phenotypes-gender imbalance and splenic marginal zone B-cell lymphoma-emerged in combination with gene dose reduction or when challenged by chronic fur mite infestation. This study highlights the role of the environment in malignancy and colitis and is consistent with Notch-dependent anti-parasite immune responses being compromised in Notch dimer-deficient animals.


Asunto(s)
Linfocitos B/inmunología , Dosificación de Gen , Corazón/embriología , Homeostasis , Intestinos/patología , Infestaciones por Ácaros/inmunología , Receptores Notch/genética , Células Madre/patología , Alelos , Animales , Secuencia de Bases , Proliferación Celular , Cromatina/metabolismo , Sulfato de Dextran , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/patología , Ratones , Ácaros/fisiología , Modelos Biológicos , Multimerización de Proteína , Receptores Notch/metabolismo , Bazo/inmunología , Esplenomegalia/inmunología , Esplenomegalia/parasitología , Células Madre/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(23): 5998-6003, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29784808

RESUMEN

Nephrogenesis concludes by the 36th week of gestation in humans and by the third day of postnatal life in mice. Extending the nephrogenic period may reduce the onset of adult renal and cardiovascular disease associated with low nephron numbers. We conditionally deleted either Mtor or Tsc1 (coding for hamartin, an inhibitor of Mtor) in renal progenitor cells. Loss of one Mtor allele caused a reduction in nephron numbers; complete deletion led to severe paucity of glomeruli in the kidney resulting in early death after birth. By contrast, loss of one Tsc1 allele from renal progenitors resulted in a 25% increase in nephron endowment with no adverse effects. Increased progenitor engraftment rates ex vivo relative to controls correlated with prolonged nephrogenesis through the fourth postnatal day. Complete loss of both Tsc1 alleles in renal progenitors led to a lethal tubular lesion. The hamartin phenotypes are not dependent on the inhibitory effect of TSC on the Mtor complex but are dependent on Raptor.


Asunto(s)
Nefronas , Organogénesis/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Nefronas/química , Nefronas/citología , Nefronas/crecimiento & desarrollo , Nefronas/fisiología , Serina-Treonina Quinasas TOR/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa
3.
Development ; 141(15): 3093-101, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053437

RESUMEN

We used a single cell RNA-seq strategy to create an atlas of gene expression patterns in the developing kidney. At several stages of kidney development, histologically uniform populations of cells give rise to multiple distinct lineages. We performed single cell RNA-seq analysis of total mouse kidneys at E11.5 and E12.5, as well as the renal vesicles at P4. We define an early stage of progenitor cell induction driven primarily by gene repression. Surprising stochastic expression of marker genes associated with differentiated cell types was observed in E11.5 progenitors. We provide a global view of the polarized gene expression already present in the renal vesicle, the first epithelial precursor of the nephron. We show that Hox gene read-through transcripts can be spliced to produce intergenic homeobox swaps. We also identify a surprising number of genes with partially degraded noncoding RNA. Perhaps most interesting, at early developmental times single cells often expressed genes related to several developmental pathways. This provides powerful evidence that initial organogenesis involves a process of multilineage priming. This is followed by a combination of gene repression, which turns off the genes associated with most possible lineages, and the activation of increasing numbers of genes driving the chosen developmental direction.


Asunto(s)
Linaje de la Célula , Riñón/embriología , Células Madre/citología , Animales , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Nefronas/embriología , Organogénesis/genética , Podocitos/citología , ARN/metabolismo , ARN no Traducido/metabolismo , Procesos Estocásticos , Factores de Tiempo
4.
Dev Biol ; 391(2): 133-46, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24780627

RESUMEN

We present a gene expression atlas of early mouse craniofacial development. Laser capture microdissection (LCM) was used to isolate cells from the principal critical microregions, whose development, differentiation and signaling interactions are responsible for the construction of the mammalian face. At E8.5, as migrating neural crest cells begin to exit the neural fold/epidermal ectoderm boundary, we examined the cranial mesenchyme, composed of mixed neural crest and paraxial mesoderm cells, as well as cells from adjacent neuroepithelium. At E9.5 cells from the cranial mesenchyme, overlying olfactory placode/epidermal ectoderm, and underlying neuroepithelium, as well as the emerging mandibular and maxillary arches were sampled. At E10.5, as the facial prominences form, cells from the medial and lateral prominences, the olfactory pit, multiple discrete regions of underlying neuroepithelium, the mandibular and maxillary arches, including both their mesenchymal and ectodermal components, as well as Rathke's pouch, were similarly sampled and profiled using both microarray and RNA-seq technologies. Further, we performed single cell studies to better define the gene expression states of the early E8.5 pioneer neural crest cells and paraxial mesoderm. Taken together, and analyzable by a variety of biological network approaches, these data provide a complementing and cross validating resource capable of fueling discovery of novel compartment specific markers and signatures whose combinatorial interactions of transcription factors and growth factors/receptors are responsible for providing the master genetic blueprint for craniofacial development.


Asunto(s)
Huesos Faciales/embriología , Regulación del Desarrollo de la Expresión Génica , Desarrollo Maxilofacial , Mesodermo/embriología , Animales , Secuencia de Bases , Diferenciación Celular , Movimiento Celular , Embrión de Mamíferos/citología , Cara/embriología , Expresión Génica , Perfilación de la Expresión Génica , Captura por Microdisección con Láser , Mesodermo/citología , Ratones , Cresta Neural/citología , Cresta Neural/embriología , Análisis de Secuencia de ARN , Transducción de Señal , Cráneo/embriología
5.
BMC Nephrol ; 16: 71, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25968128

RESUMEN

BACKGROUND: Mutations in several genes expressed in podocytes, including Cd2ap, have been associated with focal segmental glomerulosclerosis in humans. Mutant mouse models provide an opportunity to better understand the molecular pathology that drives these diseases. METHODS: In this report we use a battery of transgenic-GFP mice to facilitate the purification of all three major cell types of the glomerulus from Cd2ap mutant mice. Both microarrays and RNA-seq were used to characterize the gene expression profiles of the podocytes, mesangial cells and endothelial cells, providing a global dual platform cross-validating dataset. RESULTS: The mesangial cells showed increased expression of profibrotic factors, including thrombospondin, Tgfb2 and Tgfb3, as well as the angiogenesis factor Vegf. They also showed upregulation of protective genes, including Aldh1a2, involved in retinoic acid synthesis and Decorin, a Tgfb antagonist. Of interest, the mesangial cells also showed significant expression of Wt1, which has generally been considered podocyte specific. The Cd2ap mutant podocytes showed upregulation of proteases as well as genes involved in muscle and vasculature development and showed a very strong gene expression signature indicating programmed cell death. Endothelial cells showed increased expression of the leukocyte adhesion associated factors Vcam1 and Sele, as well as Midkine (promoting angiogenesis), endothelin and many genes responsive to cytokines and interferons. CONCLUSIONS: This study provides a comprehensive analysis of the changing properties of the three cell types of the glomerulus in Cd2ap mutants, identifying activated and repressed pathways and responsible genes, thereby delivering a deeper molecular understanding of this genetic disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/genética , Células Endoteliales/metabolismo , Glomeruloesclerosis Focal y Segmentaria/genética , Células Mesangiales/metabolismo , Podocitos/metabolismo , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Decorina/genética , Decorina/metabolismo , Modelos Animales de Enfermedad , Selectina E/genética , Selectina E/metabolismo , Endotelinas/genética , Endotelinas/metabolismo , Perfilación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Ratones , Ratones Transgénicos , Midkina , Mutación , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Retinal-Deshidrogenasa , Trombospondinas/genética , Trombospondinas/metabolismo , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas WT1
6.
Dev Biol ; 381(2): 312-23, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23872235

RESUMEN

Much of the bone, cartilage and smooth muscle of the vertebrate face is derived from neural crest (NC) cells. During craniofacial development, the anterior neural ridge (ANR) and olfactory pit (OP) signaling centers are responsible for driving the outgrowth, survival, and differentiation of NC populated facial prominences, primarily via FGF. While much is known about the functional importance of signaling centers, relatively little is understood of how these signaling centers are made and maintained. In this report we describe a dramatic craniofacial malformation in mice mutant for the zinc finger transcription factor gene Sp8. At E14.5 they show facial prominences that are reduced in size and underdeveloped, giving an almost faceless phenotype. At later times they show severe midline defects, excencephaly, hyperterlorism, cleft palate, and a striking loss of many NC and paraxial mesoderm derived cranial bones. Sp8 expression was primarily restricted to the ANR and OP regions during craniofacial development. Analysis of an extensive series of conditional Sp8 mutants confirmed the critical role of Sp8 in signaling centers, and not directly in the NC and paraxial mesoderm cells. The NC cells of the Sp8 mutants showed increased levels of apoptosis and decreased cell proliferation, thereby explaining the reduced sizes of the facial prominences. Perturbed gene expression in the Sp8 mutants was examined by laser capture microdissection coupled with microarrays, as well as in situ hybridization and immunostaining. The most dramatic differences included striking reductions in Fgf8 and Fgf17 expression in the ANR and OP signaling centers. We were also able to achieve genetic and pharmaceutical partial rescue of the Sp8 mutant phenotype by reducing Sonic Hedgehog (SHH) signaling. These results show that Sp8 primarily functions to promote Fgf expression in the ANR and OP signaling centers that drive the survival, proliferation, and differentiation of the NC and paraxial mesoderm that make the face.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Apoptosis , Cartílago/embriología , Cartílago/metabolismo , Cartílago/patología , Proliferación Celular , Anomalías Craneofaciales/embriología , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Cara/anomalías , Cara/embriología , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Ratones Transgénicos , Mutación , Cresta Neural/embriología , Cresta Neural/metabolismo , Cresta Neural/patología , Fenotipo , Embarazo , Factores de Transcripción/genética
7.
Development ; 138(13): 2845-53, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21652655

RESUMEN

The GenitoUrinary Development Molecular Anatomy Project (GUDMAP) is an international consortium working to generate gene expression data and transgenic mice. GUDMAP includes data from large-scale in situ hybridisation screens (wholemount and section) and microarray gene expression data of microdissected, laser-captured and FACS-sorted components of the developing mouse genitourinary (GU) system. These expression data are annotated using a high-resolution anatomy ontology specific to the developing murine GU system. GUDMAP data are freely accessible at www.gudmap.org via easy-to-use interfaces. This curated, high-resolution dataset serves as a powerful resource for biologists, clinicians and bioinformaticians interested in the developing urogenital system. This paper gives examples of how the data have been used to address problems in developmental biology and provides a primer for those wishing to use the database in their own research.


Asunto(s)
Bases de Datos Genéticas , Internet , Sistema Urogenital/metabolismo , Animales , Humanos , Ratones , Programas Informáticos , Sistema Urogenital/crecimiento & desarrollo
8.
Pediatr Nephrol ; 29(4): 581-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23996451

RESUMEN

Changing gene expression patterns is the essential driver of developmental processes. Growth factors, micro-RNAs, long intergenic noncoding RNAs, and epigenetic marks, such as DNA methylation and histone modifications, all work by impacting gene expression. The key features of developing cells, including their ability to communicate with others, are defined primarily by their gene-expression profiles. It is therefore clear that a gene-expression atlas of the developing kidney can provide a useful tool for the developmental nephrology research community. Toward this end, the GenitoUrinary Development Molecular Anatomy Project (GUDMAP) consortium has worked to create an atlas of the changing gene-expression patterns that drive kidney development. In this article, the global gene-expression profiling strategies of GUDMAP are reviewed. The initial work used laser-capture microdissection to purify multiple compartments of the developing kidney, including cap mesenchyme, renal vesicle, S-shaped bodies, proximal tubules, and more, which were then gene-expression profiled using microarrays. Resolution of the atlas was then improved by using transgenic mice with specific cell types labeled with green fluorescent protein (GFP), allowing their purification and profiling. In addition, RNA-Seq replaced microarrays. Currently, the atlas is being pushed to the single-cell resolution using microfluidic approaches that allow high-throughput RNA-Seq analysis of hundreds of individual cells. Results can identify novel types of cells and define interesting heterogeneities present within cell populations.


Asunto(s)
Atlas como Asunto , Perfilación de la Expresión Génica , Riñón/embriología , Humanos , Transcriptoma
9.
Dev Biol ; 368(1): 4-17, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22664176

RESUMEN

During kidney development the cap mesenchyme progenitor cells both self renew and differentiate into nephrons. The balance between renewal and differentiation determines the final nephron count, which is of considerable medical importance. An important goal is to create a precise genetic definition of the early differentiation of cap mesenchyme progenitors. We used RNA-Seq to transcriptional profile the cap mesenchyme progenitors and their first epithelial derivative, the renal vesicles. The results provide a global view of the changing gene expression program during this key period, defining expression levels for all transcription factors, growth factors, and receptors. The RNA-Seq was performed using two different biochemistries, with one examining only polyadenylated RNA and the other total RNA. This allowed the analysis of noncanonical transcripts, which for many genes were more abundant than standard exonic RNAs. Since a large fraction of enhancers are now known to be transcribed the results also provide global maps of potential enhancers. Further, the RNA-Seq data defined hundreds of novel splice patterns and large numbers of new genes. Particularly striking was the extensive sense/antisense transcription and changing RNA processing complexities of the Hox clusters.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Riñón/metabolismo , ARN/genética , Análisis de Secuencia de ARN/métodos , Animales , Proteínas de Homeodominio/genética , Riñón/citología , Riñón/crecimiento & desarrollo , Ratones , Ratones Transgénicos , Familia de Multigenes , Nefronas/citología , Nefronas/crecimiento & desarrollo , Nefronas/metabolismo , Organogénesis/genética , Isoformas de Proteínas/genética , Procesamiento Postranscripcional del ARN , Empalme del ARN , Reproducibilidad de los Resultados
10.
BMC Nephrol ; 13: 70, 2012 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-22839765

RESUMEN

BACKGROUND: Diabetic nephropathy is the leading cause of end stage renal disease. All three cell types of the glomerulus, podocytes, endothelial cells and mesangial cells, play important roles in diabetic nephropathy. In this report we used Meis1-GFP transgenic mice to purify mesangial cells from normal mice and from db/db mice, which suffer diabetic nephropathy. The purpose of the study is to better define the unique character of normal mesangial cells, and to characterize their pathogenic and protective responses during diabetic nephropathy. METHODS: Comprehensive gene expression states of the normal and diseased mesangial cells were defined with microarrays. By comparing the gene expression profiles of mesangial cells with those of multiple other renal cell types, including podocytes, endothelial cells and renal vesicles, it was possible to better define their exceptional nature, which includes smooth muscle, phagocytic and neuronal traits. RESULTS: The complete set of mesangial cell expressed transcription factors, growth factors and receptors were identified. In addition, the analysis of the mesangial cells from diabetic nephropathy mice characterized their changes in gene expression. Molecular functions and biological processes specific to diseased mesangial cells were characterized, identifying genes involved in extracellular matrix, cell division, vasculogenesis, and growth factor modulation. Selected gene changes considered of particular importance to the disease process were validated and localized within the glomuerulus by immunostaining. For example, thrombospondin, a key mediator of TGFß signaling, was upregulated in the diabetic nephropathy mesangial cells, likely contributing to fibrosis. On the other hand the decorin gene was also upregulated, and expression of this gene has been strongly implicated in the reduction of TGFß induced fibrosis. CONCLUSIONS: The results provide an important complement to previous studies examining mesangial cells grown in culture. The remarkable qualities of the mesangial cell are more fully defined in both the normal and diabetic nephropathy diseased state. New gene expression changes and biological pathways are discovered, yielding a deeper understanding of the diabetic nephropathy pathogenic process, and identifying candidate targets for the development of novel therapies.


Asunto(s)
Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Regulación de la Expresión Génica , Células Mesangiales/fisiología , Animales , Nefropatías Diabéticas/patología , Perfilación de la Expresión Génica/métodos , Ratones , Ratones Transgénicos
11.
J Am Soc Nephrol ; 22(12): 2213-25, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22034642

RESUMEN

Renin-expressing cells modulate BP, fluid-electrolyte homeostasis, and kidney development, but remarkably little is known regarding the genetic regulatory network that governs the identity of these cells. Here we compared the gene expression profiles of renin cells with most cells in the kidney at various stages of development as well as after a physiologic challenge known to induce the transformation of arteriolar smooth muscle cells into renin-expressing cells. At all stages, renin cells expressed a distinct set of genes characteristic of the renin phenotype, which was vastly different from other cell types in the kidney. For example, cells programmed to exhibit the renin phenotype expressed Akr1b7, and maturing cells expressed angiogenic factors necessary for the development of the kidney vasculature and RGS (regulator of G-protein signaling) genes, suggesting a potential relationship between renin cells and pericytes. Contrary to the plasticity of arteriolar smooth muscle cells upstream from the glomerulus, which can transiently acquire the embryonic phenotype in the adult under physiologic stress, the adult juxtaglomerular cell always possessed characteristics of both smooth muscle and renin cells. Taken together, these results identify the gene expression profile of renin-expressing cells at various stages of maturity, and suggest that juxtaglomerular cells maintain properties of both smooth muscle and renin-expressing cells, likely to allow the rapid control of body fluids and BP through both contractile and endocrine functions.


Asunto(s)
Fenómenos Fisiológicos Celulares/genética , Riñón/citología , Renina/biosíntesis , Animales , Ratones , Transcripción Genética
12.
BMC Dev Biol ; 11: 15, 2011 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-21396121

RESUMEN

BACKGROUND: The production of nephrons suddenly ends in mice shortly after birth when the remaining cells of the multi-potent progenitor mesenchyme begin to differentiate into nephrons. We exploited this terminal wave of nephron production using both microarrays and RNA-Seq to serially evaluate gene transcript levels in the progenitors. This strategy allowed us to define the changing gene expression states following induction and the onset of differentiation after birth. RESULTS: Microarray and RNA-Seq studies of the progenitors detected a change in the expression profiles of several classes of genes early after birth. One functional class, a class of genes associated with cellular proliferation, was activated. Analysis of proliferation with a nucleotide analog demonstrated in vivo that entry into the S-phase of the cell cycle preceded increases in transcript levels of genetic markers of differentiation. Microarrays and RNA-Seq also detected the onset of expression of markers of differentiation within the population of progenitors prior to detectable Six2 repression. Validation by in situ hybridization demonstrated that the markers were expressed in a subset of Six2 expressing progenitors. Finally, the studies identified a third set of genes that provide indirect evidence of an altered cellular microenvironment of the multi-potential progenitors after birth. CONCLUSIONS: These results demonstrate that Six2 expression is not sufficient to suppress activation of genes associated with growth and differentiation of nephrons. They also better define the sequence of events after induction and suggest mechanisms contributing to the rapid end of nephron production after birth in mice.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Nefronas/crecimiento & desarrollo , Nefronas/metabolismo , Proteínas Nucleares/genética , Transactivadores/genética , Factores de Transcripción/genética , Animales , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Glucólisis , Proteínas Fluorescentes Verdes , Hibridación in Situ , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Nefronas/citología , ARN/genética , Análisis de Secuencia de ARN , Células Madre/metabolismo
13.
Pediatr Nephrol ; 26(9): 1469-78, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21336810

RESUMEN

Thousands of genes show differential expression patterns during kidney development, suggesting that the genetic program driving this process is complex. While great progress has been made in defining the outline of the genetic basis of nephrogenesis, it is clear that much remains to be learned. A global atlas of the gene expression profiles of the multiple elements of the developing kidney would allow the identification of novel growth factor-receptor interactions, identify additional molecular markers of distinct components, facilitate the generation of compartment specific GFP-CRE transgenic mouse tools, lend insights into the genetic regulatory circuits governing nephron formation, and fully characterize the waves of gene expression that impel nephrogenesis. Both microarrays and next generation deep sequencing of cDNA libraries can be used to define comprehensive, sensitive, and quantitative gene expression profiles. In addition, laser capture microdissection and transgenic GFP mice can be used to isolate specific compartments and pure cell types from the developing kidney. Advancing technologies are even allowing robust gene expression profiling of single cells. The final goal is the production of an exquisitely detailed atlas of the gene expression program that drives kidney development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/embriología , Animales , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Organogénesis/genética
14.
Nature ; 434(7033): 658-62, 2005 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-15800627

RESUMEN

Mitochondria play a critical role in mediating both apoptotic and necrotic cell death. The mitochondrial permeability transition (mPT) leads to mitochondrial swelling, outer membrane rupture and the release of apoptotic mediators. The mPT pore is thought to consist of the adenine nucleotide translocator, a voltage-dependent anion channel, and cyclophilin D (the Ppif gene product), a prolyl isomerase located within the mitochondrial matrix. Here we generated mice lacking Ppif and mice overexpressing cyclophilin D in the heart. Ppif null mice are protected from ischaemia/reperfusion-induced cell death in vivo, whereas cyclophilin D-overexpressing mice show mitochondrial swelling and spontaneous cell death. Mitochondria isolated from the livers, hearts and brains of Ppif null mice are resistant to mitochondrial swelling and permeability transition in vitro. Moreover, primary hepatocytes and fibroblasts isolated from Ppif null mice are largely protected from Ca2+-overload and oxidative stress-induced cell death. However, Bcl-2 family member-induced cell death does not depend on cyclophilin D, and Ppif null fibroblasts are not protected from staurosporine or tumour-necrosis factor-alpha-induced death. Thus, cyclophilin D and the mitochondrial permeability transition are required for mediating Ca2+- and oxidative damage-induced cell death, but not Bcl-2 family member-regulated death.


Asunto(s)
Ciclofilinas/deficiencia , Mitocondrias/metabolismo , Mitocondrias/patología , Adenoviridae/genética , Animales , Atractilósido/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Encéfalo/citología , Calcio/metabolismo , Calcio/farmacología , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Peptidil-Prolil Isomerasa F , Ciclofilinas/genética , Ciclofilinas/metabolismo , Citocromos c/metabolismo , Fibroblastos , Eliminación de Gen , Peróxido de Hidrógeno/farmacología , Hígado/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Dilatación Mitocondrial/efectos de los fármacos , Dilatación Mitocondrial/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2
15.
Nat Commun ; 12(1): 6332, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732708

RESUMEN

Mammalian nephron endowment is determined by the coordinated cessation of nephrogenesis in independent niches. Here we report that translatome analysis in Tsc1+/- nephron progenitor cells from mice with elevated nephron numbers reveals how differential translation of Wnt antagonists over agonists tips the balance between self-renewal and differentiation. Wnt agonists are poorly translated in young niches, resulting in an environment with low R-spondin and high Fgf20 promoting self-renewal. In older niches we find increased translation of Wnt agonists, including R-spondin and the signalosome-promoting Tmem59, and low Fgf20, promoting differentiation. This suggests that the tipping point for nephron progenitor exit from the niche is controlled by the gradual increase in stability and possibly clustering of Wnt/Fzd complexes in individual cells, enhancing the response to ureteric bud-derived Wnt9b inputs and driving synchronized differentiation. As predicted by these findings, removing one Rspo3 allele in nephron progenitors delays cessation and increases nephron numbers in vivo.


Asunto(s)
Organogénesis/fisiología , Percepción/fisiología , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Diferenciación Celular , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio , Riñón/citología , Riñón/patología , Masculino , Proteínas de la Membrana , Ratones , Nefronas/citología , Proteínas del Tejido Nervioso , Nicho de Células Madre , Células Madre/citología , Factores de Transcripción/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Vía de Señalización Wnt
16.
Dev Biol ; 332(2): 273-86, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19501082

RESUMEN

While nephron formation is known to be initiated by a mesenchyme-to-epithelial transition of the cap mesenchyme to form a renal vesicle (RV), the subsequent patterning of the nephron and fusion with the ureteric component of the kidney to form a patent contiguous uriniferous tubule has not been fully characterized. Using dual section in situ hybridization (SISH)/immunohistochemistry (IHC) we have revealed distinct distal/proximal patterning of Notch, BMP and Wnt pathway components within the RV stage nephron. Quantitation of mitoses and Cyclin D1 expression indicated that cell proliferation was higher in the distal RV, reflecting the differential developmental programs of the proximal and distal populations. A small number of RV genes were also expressed in the early connecting segment of the nephron. Dual ISH/IHC combined with serial section immunofluorescence and 3D reconstruction revealed that fusion occurs between the late RV and adjacent ureteric tip via a process that involves loss of the intervening ureteric epithelial basement membrane and insertion of cells expressing RV markers into the ureteric tip. Using Six2-eGFPCre x R26R-lacZ mice, we demonstrate that these cells are derived from the cap mesenchyme and not the ureteric epithelium. Hence, both nephron patterning and patency are evident at the late renal vesicle stage.


Asunto(s)
Proliferación Celular , Riñón/anatomía & histología , Riñón/embriología , Mesodermo/fisiología , Morfogénesis/fisiología , Nefronas/embriología , Uréter , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Calbindinas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Epitelio/fisiología , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Riñón/fisiología , Proteínas con Homeodominio LIM , Laminina/genética , Laminina/metabolismo , Ratones , Nefronas/anatomía & histología , Nefronas/fisiología , Embarazo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Factores de Transcripción , Uréter/anatomía & histología , Uréter/embriología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
17.
Circ Res ; 101(3): 313-21, 2007 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-17569887

RESUMEN

The cardiac extracellular matrix is a dynamic structural support network that is both influenced by, and a regulator of, pathological remodeling and hypertrophic growth. In response to pathologic insults, the adult heart reexpresses the secreted extracellular matrix protein periostin (Pn). Here we show that Pn is critically involved in regulating the cardiac hypertrophic response, interstitial fibrosis, and ventricular remodeling following long-term pressure overload stimulation and myocardial infarction. Mice lacking the gene encoding Pn (Postn) were more prone to ventricular rupture in the first 10 days after a myocardial infarction, but surviving mice showed less fibrosis and better ventricular performance. Pn(-/-) mice also showed less fibrosis and hypertrophy following long-term pressure overload, suggesting an intimate relationship between Pn and the regulation of cardiac remodeling. In contrast, inducible overexpression of Pn in the heart protected mice from rupture following myocardial infarction and induced spontaneous hypertrophy with aging. With respect to a mechanism underlying these alterations, Pn(-/-) hearts showed an altered molecular program in fibroblast function. Indeed, fibroblasts isolated from Pn(-/-) hearts were less effective in adherence to cardiac myocytes and were characterized by a dramatic alteration in global gene expression (7% of all genes). These are the first genetic data detailing the function of Pn in the adult heart as a regulator of cardiac remodeling and hypertrophy.


Asunto(s)
Cardiomegalia/fisiopatología , Moléculas de Adhesión Celular/fisiología , Remodelación Ventricular/fisiología , Envejecimiento/patología , Animales , Cardiomegalia/etiología , Adhesión Celular , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cicatriz/etiología , Cicatriz/patología , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Granulocitos/patología , Rotura Cardíaca/etiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/patología , Presión , Proteínas Recombinantes de Fusión/fisiología , Regulación hacia Arriba
18.
PLoS One ; 14(8): e0216261, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31461442

RESUMEN

Focal segmental glomerulosclerosis is a major cause of end stage renal disease. Many patients prove unresponsive to available therapies. An improved understanding of the molecular basis of the disease process could provide insights leading to novel therapeutic approaches. In this study we carried out an RNA-seq analysis of the altered gene expression patterns of podocytes, mesangial cells and glomerular endothelial cells of the bigenic Cd2ap+/-, Fyn-/- mutant mouse model of FSGS. In the podocytes we observed upregulation of many genes related to the Tgfß family/pathway, including Gdnf, Tgfß1, Tgfß2, Snai2, Vegfb, Bmp4, and Tnc. The mutant podocytes also showed upregulation of Acta2, a marker of smooth muscle and associated with myofibroblasts, which are implicated in driving fibrosis. GO analysis of the podocyte upregulated genes identified elevated protein kinase activity, increased expression of growth factors, and negative regulation of cell adhesion, perhaps related to the observed podocyte loss. Both podocytes and mesangial cells showed strong upregulation of aldehyde dehydrogenase genes involved in the synthesis of retinoic acid. Similarly, the Cd2ap+/-, Fyn-/- mesangial cells, as well as podocytes in other genetic models, and the glomeruli of human FSGS patients, all show upregulation of the serine protease Prss23, with the common thread suggesting important functionality. Another gene with strong upregulation in the Cd2ap+/-, Fyn-/- mutant mesangial cells as well as multiple other mutant mouse models of FSGS was thrombospondin, which activates the secreted inactive form of Tgfß. The Cd2ap+/-, Fyn-/- mutant endothelial cells showed elevated expression of genes involved in cell proliferation, angioblast migration, angiogenesis, and neovasculature, all consistent with the formation of new blood vessels in the diseased glomerulus. The resulting global definition of the perturbed molecular pathways in the three major cell types of the mutant glomerulus provide deeper understanding of the molecular pathogenic pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/genética , Células Endoteliales/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Células Mesangiales/patología , Podocitos/patología , Proteínas Proto-Oncogénicas c-fyn/genética , Animales , Modelos Animales de Enfermedad , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Ratones , Terapia Molecular Dirigida , Mutación , Fenotipo
19.
Circ Res ; 99(9): 996-1003, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-17008600

RESUMEN

G-protein receptor kinase 2 (GRK2) is 1 of 7 mammalian GRKs that phosphorylate ligand-bound 7-transmembrane receptors, causing receptor uncoupling from G proteins and potentially activating non-G-protein signaling pathways. GRK2 is unique among members of the GRK family in that its genetic ablation causes embryonic lethality. Cardiac abnormalities in GRK2 null embryos implicated GRK2 in cardiac development but prevented studies of the knockout phenotype in adult hearts. Here, we created GRK2-loxP-targeted mice and used Cre recombination to generate germline and cardiac-specific GRK2 knockouts. GRK2 deletion in the preimplantation embryo with EIIa-Cre (germline null) resulted in developmental retardation and embryonic lethality between embryonic day 10.5 (E10.5) and E11.5. At E9.5, cardiac myocyte specification and cardiac looping were normal, but ventricular development was delayed. Cardiomyocyte-specific ablation of GRK2 in the embryo with Nkx2.5-driven Cre (cardiac-specific GRK2 knockout) produced viable mice with normal heart structure, function, and cardiac gene expression. Cardiac-specific GRK2 knockout mice exhibited enhanced inotropic sensitivity to the beta-adrenergic receptor agonist isoproterenol, with impairment of normal inotropic and lusitropic tachyphylaxis, and exhibited accelerated development of catecholamine toxicity with chronic isoproterenol treatment. These findings show that cardiomyocyte autonomous GRK2 is not essential for myocardial development after cardiac specification, suggesting that embryonic developmental abnormalities may be attributable to extracardiac effects of GRK2 ablation. In the adult heart, cardiac GRK2 is a major factor regulating inotropic and lusitropic tachyphylaxis to beta-adrenergic agonist, which likely contributes to its protective effects in catecholamine cardiomyopathy.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Corazón/embriología , Miocitos Cardíacos/enzimología , Quinasas de Receptores Adrenérgicos beta/fisiología , Agonistas Adrenérgicos beta/toxicidad , Animales , Cardiomiopatías/inducido químicamente , Cardiotónicos/farmacología , Desarrollo Embrionario , Quinasa 2 del Receptor Acoplado a Proteína-G , Marcación de Gen , Genes Letales , Isoproterenol/toxicidad , Ratones , Transducción de Señal , Taquifilaxis , Quinasas de Receptores Adrenérgicos beta/genética
20.
Circ Res ; 95(12): 1200-6, 2004 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-15539635

RESUMEN

Hundreds of signaling molecules have been assigned critical roles in the pathogenesis of myocardial hypertrophy and heart failure based on cardiac phenotypes from alpha-myosin heavy chain-directed overexpression mice. Because permanent ventricular transgene expression in this system begins during a period of rapid physiological neonatal growth, resulting phenotypes are the combined consequences of transgene effects and normal trophic influences. We used temporally-defined forced gene expression to investigate synergy between postnatal physiological cardiac growth and two functionally divergent cardiomyopathic genes. Phenotype development was compared various times after neonatal (age 2 to 3 days) and adult (age 8 weeks) expression. Proapoptotic Nix caused ventricular dilation and severe contractile depression in neonates, but not adults. Myocardial apoptosis was minimal in adults, but was widespread in neonates, until it spontaneously resolved in adulthood. Unlike normal postnatal cardiac growth, concurrent left ventricular pressure overload hypertrophy did not synergize with Nix expression to cause cardiomyopathy or myocardial apoptosis. Prohypertrophic Galphaq likewise caused eccentric hypertrophy, systolic dysfunction, and pathological gene expression in neonates, but not adults. Thus, normal postnatal cardiac growth can be an essential cofactor in development of genetic cardiomyopathies, and may confound the interpretation of conventional alpha-MHC transgenic phenotypes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Corazón/crecimiento & desarrollo , Hipertrofia Ventricular Izquierda/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Coartación Aórtica/complicaciones , Apoptosis , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Cruzamientos Genéticos , Doxiciclina/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/biosíntesis , Datos de Secuencia Molecular , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transgenes/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA