Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Stem Cell ; 29(7): 1083-1101.e7, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35803227

RESUMEN

Human pluripotent stem-cell-derived organoids are models for human development and disease. We report a modified human kidney organoid system that generates thousands of similar organoids, each consisting of 1-2 nephron-like structures. Single-cell transcriptomic profiling and immunofluorescence validation highlighted patterned nephron-like structures utilizing similar pathways, with distinct morphogenesis, to human nephrogenesis. To examine this platform for therapeutic screening, the polycystic kidney disease genes PKD1 and PKD2 were inactivated by gene editing. PKD1 and PKD2 mutant models exhibited efficient and reproducible cyst formation. Cystic outgrowths could be propagated for months to centimeter-sized cysts. To shed new light on cystogenesis, 247 protein kinase inhibitors (PKIs) were screened in a live imaging assay identifying compounds blocking cyst formation but not overall organoid growth. Scaling and further development of the organoid platform will enable a broader capability for kidney disease modeling and high-throughput drug screens.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Quistes/metabolismo , Descubrimiento de Drogas , Humanos , Riñón/metabolismo , Organoides/metabolismo , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
2.
Nat Commun ; 12(1): 2382, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33888706

RESUMEN

Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteómica/métodos , Proteínas Represoras/metabolismo , Coloración y Etiquetado/métodos , Animales , Animales Modificados Genéticamente , Biotina/metabolismo , Biotinilación , Ligasas de Carbono-Nitrógeno/genética , Línea Celular , Modelos Animales de Enfermedad , Drosophila , Células Madre Embrionarias , Proteínas de Escherichia coli/genética , Femenino , Humanos , Masculino , Ratones , Ingeniería de Proteínas , Transporte de Proteínas , Proteínas Represoras/genética , Espectrometría de Masas en Tándem/métodos , Teratoma/diagnóstico , Teratoma/patología
3.
Dev Cell ; 50(1): 102-116.e6, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31265809

RESUMEN

The renal corpuscle of the kidney comprises a glomerular vasculature embraced by podocytes and supported by mesangial myofibroblasts, which ensure plasma filtration at the podocyte-generated slit diaphragm. With a spectrum of podocyte-expressed gene mutations causing chronic disease, an enhanced understanding of podocyte development and function to create relevant in vitro podocyte models is a clinical imperative. To characterize podocyte development, scRNA-seq was performed on human fetal kidneys, identifying distinct transcriptional signatures accompanying the differentiation of functional podocytes from progenitors. Interestingly, organoid-generated podocytes exhibited highly similar, progressive transcriptional profiles despite an absence of the vasculature, although abnormal gene expression was pinpointed in late podocytes. On transplantation into mice, organoid-derived podocytes recruited the host vasculature and partially corrected transcriptional profiles. Thus, human podocyte development is mostly intrinsically regulated and vascular interactions refine maturation. These studies support the application of organoid-derived podocytes to model disease and to restore or replace normal kidney functions.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Glomérulos Renales/citología , Organoides/citología , Podocitos/citología , Análisis de la Célula Individual/métodos , Células Cultivadas , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Glomérulos Renales/metabolismo , Organoides/metabolismo , Podocitos/metabolismo
4.
Dev Cell ; 45(5): 651-660.e4, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29870722

RESUMEN

Mammalian nephrons arise from a limited nephron progenitor pool through a reiterative inductive process extending over days (mouse) or weeks (human) of kidney development. Here, we present evidence that human nephron patterning reflects a time-dependent process of recruitment of mesenchymal progenitors into an epithelial nephron precursor. Progressive recruitment predicted from high-resolution image analysis and three-dimensional reconstruction of human nephrogenesis was confirmed through direct visualization and cell fate analysis of mouse kidney organ cultures. Single-cell RNA sequencing of the human nephrogenic niche provided molecular insights into these early patterning processes and predicted developmental trajectories adopted by nephron progenitor cells in forming segment-specific domains of the human nephron. The temporal-recruitment model for nephron polarity and patterning suggested by direct analysis of human kidney development provides a framework for integrating signaling pathways driving mammalian nephrogenesis.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/citología , Nefronas/citología , Organogénesis/fisiología , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Nefronas/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Factores de Tiempo
5.
J Am Soc Nephrol ; 29(3): 806-824, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449449

RESUMEN

Cellular interactions among nephron, interstitial, and collecting duct progenitors drive mammalian kidney development. In mice, Six2+ nephron progenitor cells (NPCs) and Foxd1+ interstitial progenitor cells (IPCs) form largely distinct lineage compartments at the onset of metanephric kidney development. Here, we used the method for analyzing RNA following intracellular sorting (MARIS) approach, single-cell transcriptional profiling, in situ hybridization, and immunolabeling to characterize the presumptive NPC and IPC compartments of the developing human kidney. As in mice, each progenitor population adopts a stereotypical arrangement in the human nephron-forming niche: NPCs capped outgrowing ureteric branch tips, whereas IPCs were sandwiched between the NPCs and the renal capsule. Unlike mouse NPCs, human NPCs displayed a transcriptional profile that overlapped substantially with the IPC transcriptional profile, and key IPC determinants, including FOXD1, were readily detected within SIX2+ NPCs. Comparative gene expression profiling in human and mouse Six2/SIX2+ NPCs showed broad agreement between the species but also identified species-biased expression of some genes. Notably, some human NPC-enriched genes, including DAPL1 and COL9A2, are linked to human renal disease. We further explored the cellular diversity of mesenchymal cell types in the human nephrogenic niche through single-cell transcriptional profiling. Data analysis stratified NPCs into two main subpopulations and identified a third group of differentiating cells. These findings were confirmed by section in situ hybridization with novel human NPC markers predicted through the single-cell studies. This study provides a benchmark for the mesenchymal progenitors in the human nephrogenic niche and highlights species-variability in kidney developmental programs.


Asunto(s)
Corteza Renal/embriología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Nefronas/embriología , Animales , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular , Linaje de la Célula , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Nefronas/anatomía & histología , Nefronas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
J Am Soc Nephrol ; 26(1): 81-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24904087

RESUMEN

We previously described a mesenchymal stem cell (MSC)-like population within the adult mouse kidney that displays long-term colony-forming efficiency, clonogenicity, immunosuppression, and panmesodermal potential. Although phenotypically similar to bone marrow (BM)-MSCs, kidney MSC-like cells display a distinct expression profile. FACS sorting from Hoxb7/enhanced green fluorescent protein (GFP) mice identified the collecting duct as a source of kidney MSC-like cells, with these cells undergoing an epithelial-to-mesenchymal transition to form clonogenic, long-term, self-renewing MSC-like cells. Notably, after extensive passage, kidney MSC-like cells selectively integrated into the aquaporin 2-positive medullary collecting duct when microinjected into the kidneys of neonatal mice. No epithelial integration was observed after injection of BM-MSCs. Indeed, kidney MSC-like cells retained a capacity to form epithelial structures in vitro and in vivo, and conditioned media from these cells supported epithelial repair in vitro. To investigate the origin of kidney MSC-like cells, we further examined Hoxb7(+) fractions within the kidney across postnatal development, identifying a neonatal interstitial GFP(lo) (Hoxb7(lo)) population displaying an expression profile intermediate between epithelium and interstitium. Temporal analyses with Wnt4(GCE/+):R26(tdTomato/+) mice revealed evidence for the intercalation of a Wnt4-expressing interstitial population into the neonatal collecting duct, suggesting that such intercalation may represent a normal developmental mechanism giving rise to a distinct collecting duct subpopulation. These results extend previous observations of papillary stem cell activity and collecting duct plasticity and imply a role for such cells in collecting duct formation and, possibly, repair.


Asunto(s)
Células Epiteliales/citología , Túbulos Renales Colectores/citología , Riñón/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Separación Celular , Condrocitos/citología , Colágeno/metabolismo , Perros , Transición Epitelial-Mesenquimal , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Túbulos Renales/citología , Células de Riñón Canino Madin Darby , Ratones , Osteocitos/citología , Fenotipo
7.
Biol Reprod ; 86(2): 37, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21976597

RESUMEN

The embryonic origins of ovarian granulosa cells have been a subject of debate for decades. By tamoxifen-induced lineage tracing of Foxl2-expressing cells, we show that descendants of the bipotential supporting cell precursors in the early gonad contribute granulosa cells to a specific population of follicles in the medulla of the ovary that begin to grow immediately after birth. These precursor cells arise from the proliferative ovarian surface epithelium and enter mitotic arrest prior to upregulating Foxl2. Granulosa cells that populate the cortical primordial follicles activated in adult life derive from the surface epithelium perinatally, and enter mitotic arrest at that stage. Ingression from the surface epithelium dropped to undetectable levels by Postnatal Day 7, when most surviving oocytes were individually encapsulated by granulosa cells. These findings add complexity to the standard model of sex determination in which the Sertoli and granulosa cells of the adult testis and ovary directly stem from the supporting cell precursors of the bipotential gonad.


Asunto(s)
Linaje de la Célula , Células de la Granulosa/citología , Folículo Ovárico/citología , Ovario/embriología , Animales , Diferenciación Celular , Desarrollo Embrionario , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Células de la Granulosa/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Folículo Ovárico/metabolismo , Ovario/citología
8.
Genes Dev ; 16(22): 2849-64, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12435628

RESUMEN

The hedgehog signaling pathway organizes the developing ventral neural tube by establishing distinct neural progenitor fates along the dorsoventral axis. Smoothened (Smo) is essential for all Hedgehog (Hh) signaling, and genetic inactivation of Smo cells autonomously blocks the ability of cells to transduce the Hh signal. Using a chimeric approach, we examined the behavior of Smo null mutant neural progenitor cells in the developing vertebrate spinal cord, and we show that direct Hh signaling is essential for the specification of all ventral progenitor populations. Further, Hh signaling extends into the dorsal half of the spinal cord including the intermediate Dbx expression domain. Surprisingly, in the absence of Sonic hedgehog (Shh), we observe the presence of a Smo-dependent Hh signaling activity operating in the ventral half of the spinal cord that most likely reflects Indian hedgehog (Ihh) signaling originating from the underlying gut endoderm. Comparative studies of Shh, Smo, and Gli3 single and compound mutants reveal that Hh signaling acts in part to specify neural cell identity by counteracting the repressive action of Gli3 on p0, p1, p2, and pMN formation. However, whereas these cell identities are restored in Gli3/Smo compound mutants, correct stratification of the rescued ventral cell types is lost. Thus, Hh signaling is essential for organizing ventral cell pattern, possibly through the control of differential cell affinities.


Asunto(s)
Inducción Embrionaria/fisiología , Proteínas del Tejido Nervioso , Receptores Acoplados a Proteínas G , Proteínas Represoras , Transducción de Señal , Médula Espinal/embriología , Células Madre/fisiología , Transactivadores/metabolismo , Proteínas de Xenopus , Animales , Tipificación del Cuerpo , Quimera , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Hedgehog , Factores de Transcripción de Tipo Kruppel , Mamíferos , Ratones , Ratones Mutantes , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Neuronas/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptor Smoothened , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Gli3 con Dedos de Zinc
9.
Eur J Immunol ; 32(4): 967-71, 2002 04.
Artículo en Inglés | MEDLINE | ID: mdl-11920562

RESUMEN

Thymic primordium, formed by cells derived from the endoderm, the ectoderm and the neural crest-derived mesenchyme, receive fetal liver derived lymphoid precursors. Reciprocal cell-cell interactions between thymic stromal cells and lymphoid precursors are critical in the expansion and maturation of thymocytes. Transcription factor TCF-1 is critical for the expansion of thymocytes because deletion of TCF-1 results in a significant decrease in the number of thymocytes without affecting the developmental pattern. In this report we show that Wnt-1 and Wnt-4 are expressed in the thymus and the deletion of Wnt-1 or Wnt-4 result in a substantial decrease in the number of thymocytes without affecting the pattern of maturation. Wnt-1 and Wnt-4 both regulate developing thymocytes because a double deficiency results in a significantly greater decrease of immature and mature thymocytes compared to deficiency in either Wnt-1 or Wnt-4.


Asunto(s)
Proteínas Anfibias , Proteínas Proto-Oncogénicas/fisiología , Linfocitos T/citología , Timo/citología , Proteínas de Pez Cebra , Animales , Diferenciación Celular/genética , División Celular , Supervivencia Celular , ADN Complementario/genética , Proteínas de Unión al ADN/fisiología , Proteínas del Huevo/biosíntesis , Proteínas del Huevo/genética , Proteínas Fetales/biosíntesis , Proteínas Fetales/genética , Proteínas Fetales/fisiología , Genes Letales , Factor Nuclear 1-alfa del Hepatocito , Recuento de Linfocitos , Factor de Unión 1 al Potenciador Linfoide , Ratones , Ratones Endogámicos BALB C , Biosíntesis de Proteínas , Proteínas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Células del Estroma/citología , Factor 1 de Transcripción de Linfocitos T , Timo/embriología , Factores de Transcripción/fisiología , Proteínas Wnt , Proteína Wnt1 , Proteína Wnt3 , Proteína Wnt4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA