Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Physiol Renal Physiol ; 319(3): F423-F435, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657158

RESUMEN

Cre-lox technology has revolutionized research in renal physiology by allowing site-specific genetic recombination in individual nephron segments. The distal convoluted tubule (DCT), consisting of distinct early (DCT1) and late (DCT2) segments, plays a central role in Na+ and K+ homeostasis. The only established Cre line targeting the DCT is Pvalb-Cre, which is limited by noninducibility, activity along DCT1 only, and activity in neurons. Here, we report the characterization of the first Cre line specific to the entire DCT. CRISPR/Cas9 targeting was used to introduce a tamoxifen-inducible IRES-Cre-ERT2 cassette downstream of the coding region of the Slc12a3 gene encoding the NaCl cotransporter (NCC). The resulting Slc12a3-Cre-ERT2 mice were crossed with R26R-YFP reporter mice, which revealed minimal leakiness with 6.3% of NCC-positive cells expressing yellow fluorescent protein (YFP) in the absence of tamoxifen. After tamoxifen injection, YFP expression was observed in 91.2% of NCC-positive cells and only in NCC-positive cells, revealing high recombination efficiency and DCT specificity. Crossing to R26R-TdTomato mice revealed higher leakiness (64.5%), suggesting differential sensitivity of the floxed site. Western blot analysis revealed no differences in abundances of total NCC or the active phosphorylated form of NCC in Slc12a3-Cre-ERT2 mice of either sex compared with controls. Plasma K+ and Mg2+ concentrations and thiazide-sensitive Na+ and K+ excretion did not differ in Slc12a3-Cre-ERT2 mice compared with controls when sex matched. These data suggest genetic modification had no obvious effect on NCC function. Slc12a3-Cre-ERT2 mice are the first line generated demonstrating inducible Cre recombinase activity along the entire DCT and will be a useful tool to study DCT function.


Asunto(s)
Túbulos Renales Distales/enzimología , Recombinasas/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Animales , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Recombinasas/genética , Simportadores del Cloruro de Sodio/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Tamoxifeno/farmacología
2.
J Am Soc Nephrol ; 29(3): 825-840, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449451

RESUMEN

The nephron is the functional unit of the kidney, but the mechanism of nephron formation during human development is unclear. We conducted a detailed analysis of nephron development in humans and mice by immunolabeling, and we compared human and mouse nephron patterning to describe conserved and divergent features. We created protein localization maps that highlight the emerging patterns along the proximal-distal axis of the developing nephron and benchmark expectations for localization of functionally important transcription factors, which revealed unanticipated cellular diversity. Moreover, we identified a novel nephron subdomain marked by Wnt4 expression that we fate-mapped to the proximal mature nephron. Significant conservation was observed between human and mouse patterning. We also determined the time at which markers for mature nephron cell types first emerge-critical data for the renal organoid field. These findings have conceptual implications for the evolutionary processes driving the diversity of mammalian organ systems. Furthermore, these findings provide practical insights beyond those gained with mouse and rat models that will guide in vitro efforts to harness the developmental programs necessary to build human kidney structures.


Asunto(s)
Diferenciación Celular , Nefronas/embriología , Nefronas/metabolismo , Células Madre/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Linaje de la Célula , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt4/metabolismo
3.
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
4.
J Am Soc Nephrol ; 29(3): 785-805, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29449453

RESUMEN

Human kidney function is underpinned by approximately 1,000,000 nephrons, although the number varies substantially, and low nephron number is linked to disease. Human kidney development initiates around 4 weeks of gestation and ends around 34-37 weeks of gestation. Over this period, a reiterative inductive process establishes the nephron complement. Studies have provided insightful anatomic descriptions of human kidney development, but the limited histologic views are not readily accessible to a broad audience. In this first paper in a series providing comprehensive insight into human kidney formation, we examined human kidney development in 135 anonymously donated human kidney specimens. We documented kidney development at a macroscopic and cellular level through histologic analysis, RNA in situ hybridization, immunofluorescence studies, and transcriptional profiling, contrasting human development (4-23 weeks) with mouse development at selected stages (embryonic day 15.5 and postnatal day 2). The high-resolution histologic interactive atlas of human kidney organogenesis generated can be viewed at the GUDMAP database (www.gudmap.org) together with three-dimensional reconstructions of key components of the data herein. At the anatomic level, human and mouse kidney development differ in timing, scale, and global features such as lobe formation and progenitor niche organization. The data also highlight differences in molecular and cellular features, including the expression and cellular distribution of anchor gene markers used to identify key cell types in mouse kidney studies. These data will facilitate and inform in vitro efforts to generate human kidney structures and comparative functional analyses across mammalian species.


Asunto(s)
Riñón/embriología , Riñón/metabolismo , Organogénesis , Uréter/embriología , Animales , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Edad Gestacional , Técnicas Histológicas , Humanos , Hibridación in Situ , Riñón/anatomía & histología , Ratones , Nefronas/embriología , Nefronas/metabolismo , ARN/análisis , Uréter/metabolismo
5.
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
6.
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
7.
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
8.
Open Biol ; 12(8): 220149, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35946312

RESUMEN

Organ functions are highly specialized and interdependent. Secreted factors regulate organ development and mediate homeostasis through serum trafficking and inter-organ communication. Enzyme-catalysed proximity labelling enables the identification of proteins within a specific cellular compartment. Here, we report a BirA*G3 mouse strain that enables CRE-dependent promiscuous biotinylation of proteins trafficking through the endoplasmic reticulum. When broadly activated throughout the mouse, widespread labelling of proteins was observed within the secretory pathway. Streptavidin affinity purification and peptide mapping by quantitative mass spectrometry (MS) proteomics revealed organ-specific secretory profiles and serum trafficking. As expected, secretory proteomes were highly enriched for signal peptide-containing proteins, highlighting both conventional and non-conventional secretory processes, and ectodomain shedding. Lower-abundance proteins with hormone-like properties were recovered and validated using orthogonal approaches. Hepatocyte-specific activation of BirA*G3 highlighted liver-specific biotinylated secretome profiles. The BirA*G3 mouse model demonstrates enhanced labelling efficiency and tissue specificity over viral transduction approaches and will facilitate a deeper understanding of secretory protein interplay in development, and in healthy and diseased adult states.


Asunto(s)
Modelos Genéticos , Secretoma , Animales , Biotinilación , Mamíferos , Espectrometría de Masas/métodos , Ratones , Proteómica/métodos
9.
Nat Commun ; 12(1): 3641, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131121

RESUMEN

Current kidney organoids model development and diseases of the nephron but not the contiguous epithelial network of the kidney's collecting duct (CD) system. Here, we report the generation of an expandable, 3D branching ureteric bud (UB) organoid culture model that can be derived from primary UB progenitors from mouse and human fetal kidneys, or generated de novo from human pluripotent stem cells. In chemically-defined culture conditions, UB organoids generate CD organoids, with differentiated principal and intercalated cells adopting spatial assemblies reflective of the adult kidney's collecting system. Aggregating 3D-cultured nephron progenitor cells with UB organoids in vitro results in a reiterative process of branching morphogenesis and nephron induction, similar to kidney development. Applying an efficient gene editing strategy to remove RET activity, we demonstrate genetically modified UB organoids can model congenital anomalies of kidney and urinary tract. Taken together, these platforms will facilitate an enhanced understanding of development, regeneration and diseases of the mammalian collecting duct system.


Asunto(s)
Túbulos Renales Colectores/citología , Riñón/citología , Riñón/crecimiento & desarrollo , Organogénesis/fisiología , Organoides/citología , Organoides/crecimiento & desarrollo , Uréter , Sistema Urinario/citología , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Riñón/embriología , Túbulos Renales Colectores/embriología , Masculino , Ratones , Morfogénesis , Nefronas , Organogénesis/genética , Organoides/embriología , Células Madre Pluripotentes/citología , Sistema Urinario/embriología , Sistema Urinario/crecimiento & desarrollo
10.
Dev Cell ; 56(16): 2381-2398.e6, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34428401

RESUMEN

Congenital abnormalities of the kidney and urinary tract are among the most common birth defects, affecting 3% of newborns. The human kidney forms around a million nephrons from a pool of nephron progenitors over a 30-week period of development. To establish a framework for human nephrogenesis, we spatially resolved a stereotypical process by which equipotent nephron progenitors generate a nephron anlage, then applied data-driven approaches to construct three-dimensional protein maps on anatomical models of the nephrogenic program. Single-cell RNA sequencing identified progenitor states, which were spatially mapped to the nephron anatomy, enabling the generation of functional gene networks predicting interactions within and between nephron cell types. Network mining identified known developmental disease genes and predicted targets of interest. The spatially resolved nephrogenic program made available through the Human Nephrogenesis Atlas (https://sckidney.flatironinstitute.org/) will facilitate an understanding of kidney development and disease and enhance efforts to generate new kidney structures.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Nefronas/metabolismo , Transcriptoma , Animales , Humanos , Ratones , Nefronas/citología , Nefronas/embriología , Proteoma/genética , Proteoma/metabolismo , RNA-Seq , Análisis de la Célula Individual
11.
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
12.
Dev Biol ; 324(1): 88-98, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18835385

RESUMEN

The mammalian metanephric kidney is derived from the intermediate mesoderm. In this report, we use molecular fate mapping to demonstrate that the majority of cell types within the metanephric kidney arise from an Osr1(+) population of metanephric progenitor cells. These include the ureteric epithelium of the collecting duct network, the cap mesenchyme and its nephron epithelia derivatives, the interstitial mesenchyme, vasculature and smooth muscle. Temporal fate mapping shows a progressive restriction of Osr1(+) cell fates such that at the onset of active nephrogenesis, Osr1 activity is restricted to the Six2(+) cap mesenchyme nephron progenitors. However, low-level labeling of Osr1(+) cells suggests that the specification of interstitial mesenchyme and cap mesenchyme progenitors occurs within the Osr1(+) population prior to the onset of metanephric development. Furthermore, although Osr1(+) progenitors give rise to much of the kidney, Osr1 function is only essential for the development of the nephron progenitor compartment. These studies provide new insights into the cellular origins of metanephric kidney structures and lend support to a model where Osr1 function is limited to establishing the nephron progenitor pool.


Asunto(s)
Riñón/embriología , Mesodermo/embriología , Nefronas/embriología , Células Madre/citología , Factores de Transcripción/biosíntesis , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Proteínas de Homeodominio/metabolismo , Riñón/citología , Riñón/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Mutantes , Nefronas/citología , Nefronas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Urotelio/embriología , Urotelio/metabolismo
13.
iScience ; 20: 402-414, 2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31622881

RESUMEN

Analysis of kidney disease-causing genes and pathology resulting from systemic diseases highlight the importance of the kidney's filtering system, the renal corpuscles. To elucidate the developmental processes that establish the renal corpuscle, we performed single-nucleus droplet-based sequencing of the human fetal kidney. This enabled the identification of nephron, interstitial, and vascular cell types that together generate the renal corpuscles. Trajectory analysis identified transient developmental gene expression, predicting precursors or mature podocytes express FBLN2, BMP4, or NTN4, in conjunction with recruitment, differentiation, and modeling of vascular and mesangial cell types into a functional filter. In vitro studies provide evidence that these factors exhibit angiogenic or mesangial recruiting and inductive properties consistent with a key organizing role for podocyte precursors in kidney development. Together these studies define a spatiotemporal developmental program for the primary filtration unit of the human kidney and provide novel insights into cell interactions regulating co-assembly of constituent cell types.

14.
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
15.
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
16.
PLoS One ; 12(11): e0188413, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29145476

RESUMEN

Though many methods can be used to identify cell types contained in complex tissues, most require cell disaggregation and destroy information about where cells reside in relation to their microenvironment. Here, we describe a polytomous key for cell type identification in intact sections of adult mouse prostate and prostatic urethra. The key is organized as a decision tree and initiates with one round of immunostaining for nerve, epithelial, fibromuscular/hematolymphoid, or vascular associated cells. Cell identities are recursively eliminated by subsequent staining events until the remaining pool of potential cell types can be distinguished by direct comparison to other cells. We validated our identification key using wild type adult mouse prostate and urethra tissue sections and it currently resolves sixteen distinct cell populations which include three nerve fiber types as well as four epithelial, five fibromuscular/hematolymphoid, one nerve-associated, and three vascular-associated cell types. We demonstrate two uses of this novel identification methodology. We first used the identification key to characterize prostate stromal cell type changes in response to constitutive phosphatidylinositide-3-kinase activation in prostate epithelium. We then used the key to map cell lineages in a new reporter mouse strain driven by Wnt10aem1(cre/ERT2)Amc. The identification key facilitates rigorous and reproducible cell identification in prostate tissue sections and can be expanded to resolve additional cell types as new antibodies and other resources become available.


Asunto(s)
Próstata/metabolismo , Uretra/metabolismo , Animales , Inmunohistoquímica , Masculino , Ratones , Próstata/citología , Uretra/citología
17.
Dev Cell ; 23(3): 637-51, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22902740

RESUMEN

A balance between Six2-dependent self-renewal and canonical Wnt signaling-directed commitment regulates mammalian nephrogenesis. Intersectional studies using chromatin immunoprecipitation and transcriptional profiling identified direct target genes shared by each pathway within nephron progenitors. Wnt4 and Fgf8 are essential for progenitor commitment; cis-regulatory modules flanking each gene are cobound by Six2 and ß-catenin and are dependent on conserved Lef/Tcf binding sites for activity. In vitro and in vivo analyses suggest that Six2 and Lef/Tcf factors form a regulatory complex that promotes progenitor maintenance while entry of ß-catenin into this complex promotes nephrogenesis. Alternative transcriptional responses associated with Six2 and ß-catenin cobinding events occur through non-Lef/Tcf DNA binding mechanisms, highlighting the regulatory complexity downstream of Wnt signaling in the developing mammalian kidney.


Asunto(s)
Redes Reguladoras de Genes , Proteínas de Homeodominio/metabolismo , Nefronas/citología , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Células Cultivadas , Proteínas de Homeodominio/genética , Ratones , Ratones Transgénicos , Nefronas/embriología , Nefronas/metabolismo , Factores de Transcripción/genética , beta Catenina/genética
18.
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
19.
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