Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38674106

RESUMEN

The significant heterogeneity of Wilms' tumors between different patients is thought to arise from genetic and epigenetic distortions that occur during various stages of fetal kidney development in a way that is poorly understood. To address this, we characterized the heterogeneity of alternative mRNA splicing in Wilms' tumors using a publicly available RNAseq dataset of high-risk Wilms' tumors and normal kidney samples. Through Pareto task inference and cell deconvolution, we found that the tumors and normal kidney samples are organized according to progressive stages of kidney development within a triangle-shaped region in latent space, whose vertices, or "archetypes", resemble the cap mesenchyme, the nephrogenic stroma, and epithelial tubular structures of the fetal kidney. We identified a set of genes that are alternatively spliced between tumors located in different regions of latent space and found that many of these genes are associated with the epithelial-to-mesenchymal transition (EMT) and muscle development. Using motif enrichment analysis, we identified putative splicing regulators, some of which are associated with kidney development. Our findings provide new insights into the etiology of Wilms' tumors and suggest that specific splicing mechanisms in early stages of development may contribute to tumor development in different patients.


Asunto(s)
Empalme Alternativo , Transición Epitelial-Mesenquimal , Neoplasias Renales , Tumor de Wilms , Tumor de Wilms/genética , Tumor de Wilms/patología , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Riñón/metabolismo , Riñón/patología
2.
Science ; 382(6670): 600-606, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37917714

RESUMEN

Sex determination in mammals depends on the differentiation of the supporting lineage of the gonads into Sertoli or pregranulosa cells that govern testis and ovary development, respectively. Although the Y-linked testis-determining gene Sry has been identified, the ovarian-determining factor remains unknown. In this study, we identified -KTS, a major, alternatively spliced isoform of the Wilms tumor suppressor WT1, as a key determinant of female sex determination. Loss of -KTS variants blocked gonadal differentiation in mice, whereas increased expression, as found in Frasier syndrome, induced precocious differentiation of ovaries independently of their genetic sex. In XY embryos, this antagonized Sry expression, resulting in male-to-female sex reversal. Our results identify -KTS as an ovarian-determining factor and demonstrate that its time of activation is critical in gonadal sex differentiation.


Asunto(s)
Ovario , Procesos de Determinación del Sexo , Proteínas WT1 , Animales , Femenino , Masculino , Ratones , Ovario/crecimiento & desarrollo , Procesos de Determinación del Sexo/genética , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/crecimiento & desarrollo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Isoformas de Proteínas
4.
Cell Rep Med ; 4(3): 100939, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36796366

RESUMEN

Immune checkpoint therapy (ICT) has the power to eradicate cancer, but the mechanisms that determine effective therapy-induced immune responses are not fully understood. Here, using high-dimensional single-cell profiling, we interrogate whether the landscape of T cell states in the peripheral blood predict responses to combinatorial targeting of the OX40 costimulatory and PD-1 inhibitory pathways. Single-cell RNA sequencing and mass cytometry expose systemic and dynamic activation states of therapy-responsive CD4+ and CD8+ T cells in tumor-bearing mice with expression of distinct natural killer (NK) cell receptors, granzymes, and chemokines/chemokine receptors. Moreover, similar NK cell receptor-expressing CD8+ T cells are also detected in the blood of immunotherapy-responsive cancer patients. Targeting the NK cell and chemokine receptors in tumor-bearing mice shows the functional importance of these receptors for therapy-induced anti-tumor immunity. These findings provide a better understanding of ICT and highlight the use and targeting of dynamic biomarkers on T cells to improve cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Ratones , Antígeno B7-H1 , Diferenciación Celular , Neoplasias/patología , Receptores de Quimiocina
5.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36834944

RESUMEN

Wilms' tumors are pediatric malignancies that are thought to arise from faulty kidney development. They contain a wide range of poorly differentiated cell states resembling various distorted developmental stages of the fetal kidney, and as a result, differ between patients in a continuous manner that is not well understood. Here, we used three computational approaches to characterize this continuous heterogeneity in high-risk blastemal-type Wilms' tumors. Using Pareto task inference, we show that the tumors form a triangle-shaped continuum in latent space that is bounded by three tumor archetypes with "stromal", "blastemal", and "epithelial" characteristics, which resemble the un-induced mesenchyme, the cap mesenchyme, and early epithelial structures of the fetal kidney. By fitting a generative probabilistic "grade of membership" model, we show that each tumor can be represented as a unique mixture of three hidden "topics" with blastemal, stromal, and epithelial characteristics. Likewise, cellular deconvolution allows us to represent each tumor in the continuum as a unique combination of fetal kidney-like cell states. These results highlight the relationship between Wilms' tumors and kidney development, and we anticipate that they will pave the way for more quantitative strategies for tumor stratification and classification.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Niño , Humanos , Neoplasias Renales/patología , Aprendizaje Automático no Supervisado , Riñón/patología
6.
Exp Mol Med ; 54(12): 2188-2199, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36494589

RESUMEN

The generation of conditional alleles using CRISPR technology is still challenging. Here, we introduce a Short Conditional intrON (SCON, 189 bp) that enables the rapid generation of conditional alleles via one-step zygote injection. In this study, a total of 13 SCON mouse lines were successfully generated by 2 different laboratories. SCON has conditional intronic functions in various vertebrate species, and its target insertion is as simple as CRISPR/Cas9-mediated gene tagging.


Asunto(s)
Sistemas CRISPR-Cas , Cigoto , Ratones , Animales , Sistemas CRISPR-Cas/genética , Intrones/genética , Técnicas de Inactivación de Genes
7.
Stem Cell Reports ; 16(11): 2718-2735, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34678205

RESUMEN

In Parkinson's disease (PD), substantia nigra (SN) dopaminergic (DA) neurons degenerate, while related ventral tegmental area (VTA) DA neurons remain relatively unaffected. Here, we present a methodology that directs the differentiation of mouse and human pluripotent stem cells toward either SN- or VTA-like DA lineage and models their distinct vulnerabilities. We show that the level of WNT activity is critical for the induction of the SN- and VTA-lineage transcription factors Sox6 and Otx2, respectively. Both WNT signaling modulation and forced expression of these transcription factors can drive DA neurons toward the SN- or VTA-like fate. Importantly, the SN-like lineage enriched DA cultures recapitulate the selective sensitivity to mitochondrial toxins as observed in PD, while VTA-like neuron-enriched cultures are more resistant. Furthermore, a proteomics approach led to the identification of compounds that alter SN neuronal survival, demonstrating the utility of our strategy for disease modeling and drug discovery.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Degeneración Nerviosa/genética , Enfermedad de Parkinson/genética , Células Madre Pluripotentes/metabolismo , Sustancia Negra/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Neuronas Dopaminérgicas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Modelos Neurológicos , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Células Madre Pluripotentes/citología , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Sustancia Negra/citología , Área Tegmental Ventral/citología
8.
Genes (Basel) ; 12(2)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33672414

RESUMEN

The adult mammalian kidney is a poorly regenerating organ that lacks the stem cells that could replenish functional homeostasis similarly to, e.g., skin or the hematopoietic system. Unlike a mature kidney, the embryonic kidney hosts at least three types of lineage-specific stem cells that give rise to (a) a ureter and collecting duct system, (b) nephrons, and (c) mesangial cells together with connective tissue of the stroma. Extensive interest has been raised towards these embryonic progenitor cells, which are normally lost before birth in humans but remain part of the undifferentiated nephrogenic rests in the pediatric renal cancer Wilms tumor. Here, we discuss the current understanding of kidney-specific embryonic progenitor regulation in the innate environment of the developing kidney and the types of disruptions in their balanced regulation that lead to the formation of Wilms tumor.


Asunto(s)
Desarrollo Embrionario/genética , Riñón/crecimiento & desarrollo , Organogénesis/genética , Tumor de Wilms/genética , Animales , Diferenciación Celular/genética , Niño , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Humanos , Riñón/patología , Tumor de Wilms/patología
9.
J Immunol ; 205(11): 3154-3166, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139489

RESUMEN

The proliferation, differentiation, and survival of cells of the mononuclear phagocyte system (MPS; progenitors, monocytes, macrophages, and classical dendritic cells) are controlled by signals from the M-CSF receptor (CSF1R). Cells of the MPS lineage have been identified using numerous surface markers and transgenic reporters, but none is both universal and lineage restricted. In this article, we report the development and characterization of a CSF1R reporter mouse. A FusionRed (FRed) cassette was inserted in-frame with the C terminus of CSF1R, separated by a T2A-cleavable linker. The insertion had no effect of CSF1R expression or function. CSF1R-FRed was expressed in monocytes and macrophages and absent from granulocytes and lymphocytes. In bone marrow, CSF1R-FRed was absent in lineage-negative hematopoietic stem cells, arguing against a direct role for CSF1R in myeloid lineage commitment. It was highly expressed in marrow monocytes and common myeloid progenitors but significantly lower in granulocyte-macrophage progenitors. In sections of bone marrow, CSF1R-FRed was also detected in osteoclasts, CD169+ resident macrophages, and, consistent with previous mRNA analysis, in megakaryocytes. In lymphoid tissues, CSF1R-FRed highlighted diverse MPS populations, including classical dendritic cells. Whole mount imaging of nonlymphoid tissues in mice with combined CSF1R-FRed/Csf1r-EGFP confirmed the restriction of CSF1R expression to MPS cells. The two markers highlight the remarkable abundance and regular distribution of tissue MPS cells, including novel macrophage populations within tendon and skeletal muscle and underlying the mesothelial/serosal/capsular surfaces of every major organ. The CSF1R-FRed mouse provides a novel reporter with exquisite specificity for cells of the MPS.


Asunto(s)
Biomarcadores/metabolismo , Sistema Mononuclear Fagocítico/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Animales , Diferenciación Celular/fisiología , Células Dendríticas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/metabolismo , Músculo Esquelético/metabolismo , ARN Mensajero/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Tendones/metabolismo
10.
J Am Soc Nephrol ; 31(10): 2278-2291, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32651222

RESUMEN

BACKGROUND: During mammalian kidney development, nephron progenitors undergo a mesenchymal-to-epithelial transition and eventually differentiate into the various tubular segments of the nephron. Recently, Drop-seq single-cell RNA sequencing technology for measuring gene expression from thousands of individual cells identified the different cell types in the developing kidney. However, that analysis did not include the additional layer of heterogeneity that alternative mRNA splicing creates. METHODS: Full transcript length single-cell RNA sequencing characterized the transcriptomes of 544 individual cells from mouse embryonic kidneys. RESULTS: Gene expression levels measured with full transcript length single-cell RNA sequencing identified each cell type. Further analysis comprehensively characterized splice isoform switching during the transition between mesenchymal and epithelial cellular states, which is a key transitional process in kidney development. The study also identified several putative splicing regulators, including the genes Esrp1/2 and Rbfox1/2. CONCLUSIONS: Discovery of the sets of genes that are alternatively spliced as the fetal kidney mesenchyme differentiates into tubular epithelium will improve our understanding of the molecular mechanisms that drive kidney development.


Asunto(s)
Riñón/embriología , Mesodermo/embriología , Organogénesis/genética , Urotelio/embriología , Animales , Técnicas de Cultivo de Célula , Ratones , Isoformas de ARN , Análisis de Secuencia de ARN
11.
J Bone Miner Res ; 35(2): 357-367, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31596966

RESUMEN

Arterial calcification is an important hallmark of cardiovascular disease and shares many similarities with skeletal mineralization. The bone-specific protein osteocalcin (OCN) is an established marker of vascular smooth muscle cell (VSMC) osteochondrogenic transdifferentiation and a known regulator of glucose metabolism. However, the role of OCN in controlling arterial calcification is unclear. We hypothesized that OCN regulates calcification in VSMCs and sought to identify the underpinning signaling pathways. Immunohistochemistry revealed OCN co-localization with VSMC calcification in human calcified carotid artery plaques. Additionally, 3 mM phosphate treatment stimulated OCN mRNA expression in cultured VSMCs (1.72-fold, p < 0.001). Phosphate-induced calcification was blunted in VSMCs derived from OCN null mice (Ocn -/- ) compared with cells derived from wild-type (WT) mice (0.37-fold, p < 0.001). Ocn -/- VSMCs showed reduced mRNA expression of the osteogenic marker Runx2 (0.51-fold, p < 0.01) and the sodium-dependent phosphate transporter, PiT1 (0.70-fold, p < 0.001), with an increase in the calcification inhibitor Mgp (1.42-fold, p < 0.05) compared with WT. Ocn -/- VSMCs also showed reduced mRNA expression of Axin2 (0.13-fold, p < 0.001) and Cyclin D (0.71 fold, p < 0.01), markers of Wnt signaling. CHIR99021 (GSK3ß inhibitor) treatment increased calcium deposition in WT and Ocn -/- VSMCs (1 µM, p < 0.001). Ocn -/- VSMCs, however, calcified less than WT cells (1 µM; 0.27-fold, p < 0.001). Ocn -/- VSMCs showed reduced mRNA expression of Glut1 (0.78-fold, p < 0.001), Hex1 (0.77-fold, p < 0.01), and Pdk4 (0.47-fold, p < 0.001). This was accompanied by reduced glucose uptake (0.38-fold, p < 0.05). Subsequent mitochondrial function assessment revealed increased ATP-linked respiration (1.29-fold, p < 0.05), spare respiratory capacity (1.59-fold, p < 0.01), and maximal respiration (1.52-fold, p < 0.001) in Ocn -/- versus WT VSMCs. Together these data suggest that OCN plays a crucial role in arterial calcification mediated by Wnt/ß-catenin signaling through reduced maximal respiration. Mitochondrial dynamics may therefore represent a novel therapeutic target for clinical intervention. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Calcificación Vascular , Vía de Señalización Wnt , Animales , Células Cultivadas , Glucosa , Ratones , Músculo Liso Vascular , Miocitos del Músculo Liso , Osteocalcina/genética
12.
Nat Commun ; 10(1): 3215, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324781

RESUMEN

The proliferation, differentiation and survival of mononuclear phagocytes depend on signals from the receptor for macrophage colony-stimulating factor, CSF1R. The mammalian Csf1r locus contains a highly conserved super-enhancer, the fms-intronic regulatory element (FIRE). Here we show that genomic deletion of FIRE in mice selectively impacts CSF1R expression and tissue macrophage development in specific tissues. Deletion of FIRE ablates macrophage development from murine embryonic stem cells. Csf1rΔFIRE/ΔFIRE mice lack macrophages in the embryo, brain microglia and resident macrophages in the skin, kidney, heart and peritoneum. The homeostasis of other macrophage populations and monocytes is unaffected, but monocytes and their progenitors in bone marrow lack surface CSF1R. Finally, Csf1rΔFIRE/ΔFIRE mice are healthy and fertile without the growth, neurological or developmental abnormalities reported in Csf1r-/- rodents. Csf1rΔFIRE/ΔFIRE mice thus provide a model to explore the homeostatic, physiological and immunological functions of tissue-specific macrophage populations in adult animals.


Asunto(s)
Genes fms/genética , Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Eliminación de Secuencia , Animales , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/patología , Factor de Crecimiento Epidérmico , Femenino , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Monocitos/metabolismo , Fagocitosis , Células RAW 264.7 , Secuencias Reguladoras de Ácidos Nucleicos/genética
13.
Elife ; 82019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30758286

RESUMEN

The origins and functions of kidney macrophages in the adult have been explored, but their roles during development remain largely unknown. Here we characterise macrophage arrival, localisation, heterogeneity, and functions during kidney organogenesis. Using genetic approaches to ablate macrophages, we identify a role for macrophages in nephron progenitor cell clearance as mouse kidney development begins. Throughout renal organogenesis, most kidney macrophages are perivascular and express F4/80 and CD206. These macrophages are enriched for mRNAs linked to developmental processes, such as blood vessel morphogenesis. Using antibody-mediated macrophage-depletion, we show macrophages support vascular anastomoses in cultured kidney explants. We also characterise a subpopulation of galectin-3+ (Gal3+) myeloid cells within the developing kidney. Our findings may stimulate research into macrophage-based therapies for renal developmental abnormalities and have implications for the generation of bioengineered kidney tissues.


Asunto(s)
Galectina 3/genética , Riñón/crecimiento & desarrollo , Nefronas/crecimiento & desarrollo , Organogénesis/genética , Animales , Proteínas de Unión al Calcio/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Riñón/metabolismo , Lectinas Tipo C/genética , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Ratones , Nefronas/metabolismo , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/metabolismo
14.
Kidney Int ; 93(4): 903-920, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29398135

RESUMEN

The Wilms' tumor suppressor gene, WT1, encodes a zinc finger protein that regulates podocyte development and is highly expressed in mature podocytes. Mutations in the WT1 gene are associated with the development of renal failure due to the formation of scar tissue within glomeruli, the mechanisms of which are poorly understood. Here, we used a tamoxifen-based CRE-LoxP system to induce deletion of Wt1 in adult mice to investigate the mechanisms underlying evolution of glomerulosclerosis. Podocyte apoptosis was evident as early as the fourth day post-induction and increased during disease progression, supporting a role for Wt1 in mature podocyte survival. Podocyte Notch activation was evident at disease onset with upregulation of Notch1 and its transcriptional targets, including Nrarp. There was repression of podocyte FoxC2 and upregulation of Hey2 supporting a role for a Wt1/FoxC2/Notch transcriptional network in mature podocyte injury. The expression of cleaved Notch1 and HES1 proteins in podocytes of mutant mice was confirmed in early disease. Furthermore, induction of podocyte HES1 expression was associated with upregulation of genes implicated in epithelial mesenchymal transition, thereby suggesting that HES1 mediates podocyte EMT. Lastly, early pharmacological inhibition of Notch signaling ameliorated glomerular scarring and albuminuria. Thus, loss of Wt1 in mature podocytes modulates podocyte Notch activation, which could mediate early events in WT1-related glomerulosclerosis.


Asunto(s)
Glomerulonefritis/metabolismo , Podocitos/metabolismo , Receptor Notch1/metabolismo , Proteínas Represoras/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/patología , Péptidos y Proteínas de Señalización Intracelular , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , Proteínas/genética , Proteínas/metabolismo , Receptor Notch1/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Transcripción Genética , Proteínas WT1
15.
Stem Cell Reports ; 9(4): 1152-1166, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28966119

RESUMEN

Wilms' tumor suppressor 1 (WT1) plays an important role in cell proliferation and mesenchymal-epithelial balance in normal development and disease. Here, we show that following podocyte depletion in three experimental models, and in patients with focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, WT1 increased significantly in cells of renin lineage (CoRL). In an animal model of FSGS in RenWt1fl/fl reporter mice with inducible deletion of WT1 in CoRL, CoRL proliferation and migration to the glomerulus was reduced, and glomerular disease was worse compared with wild-type mice. To become podocytes, CoRL undergo mesenchymal-to-epithelial transformation (MET), typified by reduced staining for mesenchymal markers (MYH11, SM22, αSMA) and de novo expression of epithelial markers (E-cadherin and cytokeratin18). Evidence for changes in MET markers was barely detected in RenWt1fl/fl mice. Our results show that following podocyte depletion, WT1 plays essential roles in CoRL proliferation and migration toward an adult podocyte fate.


Asunto(s)
Linaje de la Célula , Podocitos/metabolismo , Renina/genética , Proteínas WT1/genética , Animales , Biomarcadores , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Pruebas de Función Renal , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Ratones , Ratones Noqueados , Podocitos/citología , Renina/metabolismo , Proteínas WT1/metabolismo
16.
Dev Dyn ; 246(12): 1047-1056, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28929539

RESUMEN

BACKGROUND: During murine kidney development, new cortical blood vessels form and pattern in cycles that coincide with cycles of collecting duct branching and the accompanying splitting of the cap mesenchyme (nephron progenitor cell populations that "cap" collecting duct ends). At no point in the patterning cycle do blood vessels enter the cap mesenchyme. We hypothesized that the exclusion of blood vessels from the cap mesenchyme may be controlled, at least in part, by an anti-angiogenic signal expressed by the cap mesenchyme cells. RESULTS: We show that semaphorin-3f (Sema3f), a known anti-angiogenic factor, is expressed in cap mesenchymal cells and its receptor, neuropilin-2 (Nrp2), is expressed by newly forming blood vessels in the cortex of the developing kidney. We hypothesized that Sema3f/Nrp2 signaling excludes vessels from the cap mesenchyme. Genetic ablation of Sema3f and of Nrp2, however, failed to result in vessels invading the cap mesenchyme. CONCLUSIONS: Despite complementary expression patterns, our data suggest that Sema3f and Nrp2 are dispensable for the exclusion of vessels from the cap mesenchyme during kidney development. These results should provoke additional experiments to ascertain the biological significance of Sema3f/Nrp2 expression in the developing kidney. Developmental Dynamics 246:1047-1056, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Riñón , Proteínas de la Membrana/biosíntesis , Mesodermo , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Neuropilina-2/biosíntesis , Animales , Riñón/irrigación sanguínea , Riñón/embriología , Proteínas de la Membrana/genética , Mesodermo/irrigación sanguínea , Mesodermo/embriología , Ratones , Proteínas del Tejido Nervioso/genética , Neuropilina-2/genética
17.
Sci Rep ; 7(1): 3273, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28607473

RESUMEN

The renal vasculature is required for blood filtration, blood pressure regulation, and pH maintenance, as well as other specialised kidney functions. Yet, despite its importance, many aspects of its development are poorly understood. To provide a detailed spatiotemporal analysis of kidney vascularisation, we collected images of embryonic mouse kidneys at various developmental time-points. Here we describe the first stages of kidney vascularisation and demonstrate that polygonal networks of vessels (endothelial plexuses) form in cycles at the periphery of the kidney. We show that kidney vascularisation initiates at E11, when vessels connected to the embryonic circulation form a ring around the ureteric bud. From E13.5, endothelial plexuses organise around populations of cap mesenchymal and ureteric bud cells in a cyclical, predictable manner. Specifically, as the ureteric bud bifurcates, endothelia form across the bifurcation site as the cap mesenchyme splits. The plexuses are vascular, carry erythrocytes, are enclosed within a basement membrane, and can always be traced back to the renal artery. Our results are a major step towards understanding how the global architecture of the renal vasculature is achieved.


Asunto(s)
Vasos Sanguíneos/embriología , Riñón/irrigación sanguínea , Riñón/embriología , Nefronas/irrigación sanguínea , Nefronas/embriología , Organogénesis , Animales , Biomarcadores , Vasos Sanguíneos/metabolismo , Endotelio/metabolismo , Técnica del Anticuerpo Fluorescente , Membrana Basal Glomerular/metabolismo , Riñón/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Modelos Biológicos , Nefronas/metabolismo
18.
J Cell Physiol ; 232(11): 3006-3019, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28488815

RESUMEN

The transmembrane glycoprotein E11/Podoplanin (Pdpn) has been implicated in the initial stages of osteocyte differentiation. However, its precise function and regulatory mechanisms are still unknown. Due to the known embryonic lethality induced by global Pdpn deletion, we have herein explored the effect of bone-specific Pdpn knockdown on osteocyte form and function in the post-natal mouse. Extensive skeletal phenotyping of male and female 6-week-old Oc-cre;Pdpnflox/flox (cKO) mice and their Pdpnflox/flox controls (fl/fl) has revealed that Pdpn deletion significantly compromises tibial cortical bone microarchitecture in both sexes, albeit to different extents (p < 0.05). Consistent with this, we observed an increase in stiffness in female cKO mice in comparison to fl/fl mice (p < 0.01). Moreover, analysis of the osteocyte phenotype by phalloidin staining revealed a significant decrease in the dendrite volume (p < 0.001) and length (p < 0.001) in cKO mice in which deletion of Pdpn also modifies the bone anabolic loading response (p < 0.05) in comparison to age-matched fl/fl mice. Together, these data confirm a regulatory role for Pdpn in osteocyte dendrite formation and as such, in the control of osteocyte function. As the osteocyte dendritic network is known to play vital roles in regulating bone modeling/remodeling, this highlights an essential role for Pdpn in bone homeostasis.


Asunto(s)
Diferenciación Celular , Forma de la Célula , Eliminación de Gen , Glicoproteínas de Membrana/metabolismo , Osteocitos/metabolismo , Osteogénesis , Tibia/metabolismo , Animales , Femenino , Genotipo , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones Noqueados , Osteocitos/patología , Fenotipo , Transducción de Señal , Tibia/diagnóstico por imagen , Tibia/patología , Microtomografía por Rayos X
19.
Genes Dev ; 31(4): 347-352, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28289143

RESUMEN

Wilms' tumor 1 (WT1) is essential for the development and homeostasis of multiple mesodermal tissues. Despite evidence for post-transcriptional roles, no endogenous WT1 target RNAs exist. Using RNA immunoprecipitation and UV cross-linking, we show that WT1 binds preferentially to 3' untranslated regions (UTRs) of developmental targets. These target mRNAs are down-regulated upon WT1 depletion in cell culture and developing kidney mesenchyme. Wt1 deletion leads to rapid turnover of specific mRNAs. WT1 regulates reporter gene expression through interaction with 3' UTR-binding sites. Combining experimental and computational analyses, we propose that WT1 influences key developmental and disease processes in part through regulating mRNA turnover.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , ARN Mensajero/genética , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Eliminación de Gen , Riñón/citología , Mesodermo/metabolismo , Ratones , Células Madre Embrionarias de Ratones , ARN Mensajero/metabolismo
20.
Sci Rep ; 7: 45255, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28345629

RESUMEN

WT1 is a transcription factor which regulates the epithelial-mesenchymal balance during embryonic development and, if mutated, can lead to the formation of Wilms' tumour, the most common paediatric kidney cancer. Its expression has also been reported in several adult tumour types, including breast cancer, and usually correlates with poor outcome. However, published data is inconsistent and the role of WT1 in this malignancy remains unclear. Here we provide a complete study of WT1 expression across different breast cancer subtypes as well as isoform specific expression analysis. Using in vitro cell lines, clinical samples and publicly available gene expression datasets, we demonstrate that WT1 plays a role in regulating the epithelial-mesenchymal balance of breast cancer cells and that WT1-expressing tumours are mainly associated with a mesenchymal phenotype. WT1 gene expression also correlates with CYP3A4 levels and is associated with poorer response to taxane treatment. Our work is the first to demonstrate that the known association between WT1 expression in breast cancer and poor prognosis is potentially due to cancer-related epithelial-to-mesenchymal transition (EMT) and poor chemotherapy response.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP3A/metabolismo , Taxoides/uso terapéutico , Proteínas WT1/genética , Proteínas WT1/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Docetaxel , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Mutación , Pronóstico , Taxoides/farmacología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA