Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 432
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39250420

RESUMEN

In vivo and in vitro studies argue that concentration-dependent Wnt signaling regulates mammalian nephron progenitor cell (NPC) programs. Canonical Wnt signaling is regulated through the stabilization of ß-catenin, a transcriptional co-activator when complexed with Lef/Tcf DNA-binding partners. Using the GSK3ß inhibitor CHIR99021 (CHIR) to block GSK3ß-dependent destruction of ß-catenin, we examined dose-dependent responses to ß-catenin in mouse NPCs, using mRNA transduction to modify gene expression. Low CHIR-dependent proliferation of NPCs was blocked on ß-catenin removal, with evidence of NPCs arresting at the G2-M transition. While NPC identity was maintained following ß-catenin removal, mRNA-seq identified low CHIR and ß-catenin dependent genes. High CHIR activated nephrogenesis. Nephrogenic programming was dependent on Lef/Tcf factors and ß-catenin transcriptional activity. Molecular and cellular features of early nephrogenesis were driven in the absence of CHIR by a mutated stabilized form of ß-catenin. Chromatin association studies indicate low and high CHIR response genes are likely direct targets of canonical Wnt transcriptional complexes. Together, these studies provide evidence for concentration-dependent Wnt signaling in the regulation of NPCs and provide new insight into Wnt targets initiating mammalian nephrogenesis.


Asunto(s)
Nefronas , Células Madre , Vía de Señalización Wnt , beta Catenina , Animales , Nefronas/metabolismo , Nefronas/citología , beta Catenina/metabolismo , Ratones , Células Madre/metabolismo , Células Madre/citología , Pirimidinas/farmacología , Piridinas/farmacología , Regulación del Desarrollo de la Expresión Génica , Proliferación Celular , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Organogénesis/genética , Transcripción Genética
2.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39344436

RESUMEN

In the developing mammalian kidney, nephron formation is initiated by a subset of nephron progenitor cells (NPCs). Wnt input activates a ß-catenin (Ctnnb1)-driven, transcriptional nephrogenic program and the mesenchymal to epithelial transition (MET) of NPCs. Using an in vitro mouse NPC culture model, we observed that activation of the Wnt pathway results in the aggregation of induced NPCs, which is an initiating step in the MET program. Genetic removal showed aggregation was dependent on ß-catenin. Modulating extracellular Ca2+ levels showed cell-cell contacts were Ca2+ dependent, suggesting a role for cadherin (Cdh)-directed cell adhesion. Molecular analysis identified Cdh2, Cdh4 and Cdh11 in NPCs, and the ß-catenin directed upregulation of Cdh3 and Cdh4 accompanying the MET of induced NPCs. Mutational analysis of ß-catenin supported a role for a Lef/Tcf-ß-catenin-mediated transcriptional response in the cell aggregation process. Genetic removal of all four cadherins, and independent removal of α-catenin or of ß-catenin-α-catenin interactions, abolished aggregation, but not the inductive response to Wnt pathway activation. These findings, and data in an accompanying article highlight the role of ß-catenin in linking transcriptional programs to the morphogenesis of NPCs in mammalian nephrogenesis.


Asunto(s)
Cadherinas , Agregación Celular , Transición Epitelial-Mesenquimal , Nefronas , Células Madre , Vía de Señalización Wnt , beta Catenina , Animales , Cadherinas/metabolismo , Cadherinas/genética , Nefronas/metabolismo , Nefronas/citología , Células Madre/metabolismo , Células Madre/citología , beta Catenina/metabolismo , beta Catenina/genética , Ratones , Transición Epitelial-Mesenquimal/genética , Adhesión Celular , Proteínas Wnt/metabolismo , Proteínas Wnt/genética , Células Cultivadas
3.
Proc Natl Acad Sci U S A ; 121(39): e2404586121, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39292750

RESUMEN

Developmental biology-inspired strategies for tissue-building have extraordinary promise for regenerative medicine, spurring interest in the relationship between cell biophysical properties and morphological transitions. However, mapping gene or protein expression data to cell biophysical properties to physical morphogenesis remains challenging with current techniques. Here, we present multiplexed adhesion and traction of cells at high yield (MATCHY). MATCHY advances the multiplexing and throughput capabilities of existing traction force and cell-cell adhesion assays using microfabrication and a semiautomated computation scheme with machine learning-driven cell segmentation. Both biophysical assays are coupled with serial downstream immunofluorescence to extract cell type/signaling state information. MATCHY is especially suited to complex primary tissue-, organoid-, or biopsy-derived cell mixtures since it does not rely on a priori knowledge of cell surface markers, cell sorting, or use of lineage-specific reporter animals. We first validate MATCHY on canine kidney epithelial cells engineered for rearranged during transfection (RET) tyrosine kinase expression and quantify a relationship between downstream signaling and cell traction. We then use MATCHY to create a biophysical atlas of mouse embryonic kidney primary cells and identify distinct biophysical states along the nephron differentiation trajectory. Our data complement expression-level knowledge of adhesion molecule changes that accompany nephron differentiation with quantitative biophysical information. These data reveal an "energetic ratchet" that accounts for spatial trends in nephron progenitor cell condensation as they differentiate into early nephron structures, which we validate through agent-based computational simulation. MATCHY offers semiautomated cell biophysical characterization at >10,000-cell throughput, an advance benefiting fundamental studies and new synthetic tissue strategies for regenerative medicine.


Asunto(s)
Adhesión Celular , Nefronas , Animales , Perros , Nefronas/metabolismo , Nefronas/citología , Ratones , Diferenciación Celular , Células de Riñón Canino Madin Darby , Transducción de Señal
4.
Genes Dev ; 32(13-14): 903-908, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950491

RESUMEN

Loss of function of the DIS3L2 exoribonuclease is associated with Wilms tumor and the Perlman congenital overgrowth syndrome. LIN28, a Wilms tumor oncoprotein, triggers the DIS3L2-mediated degradation of the precursor of let-7, a microRNA that inhibits Wilms tumor development. These observations have led to speculation that DIS3L2-mediated tumor suppression is attributable to let-7 regulation. Here we examine new DIS3L2-deficient cell lines and mouse models, demonstrating that DIS3L2 loss has no effect on mature let-7 levels. Rather, analysis of Dis3l2-null nephron progenitor cells, a potential cell of origin of Wilms tumors, reveals up-regulation of Igf2, a growth-promoting gene strongly associated with Wilms tumorigenesis. These findings nominate a new potential mechanism underlying the pathology associated with DIS3L2 deficiency.


Asunto(s)
Exorribonucleasas/genética , Macrosomía Fetal/genética , Factor II del Crecimiento Similar a la Insulina/genética , Regulación hacia Arriba , Tumor de Wilms/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , MicroARNs/genética , Mutación , Nefronas/citología , Nefronas/fisiopatología , Células Madre
5.
Nucleic Acids Res ; 50(18): 10343-10359, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36130284

RESUMEN

Eya1 is critical for establishing and maintaining nephron progenitor cells (NPCs). It belongs to a family of proteins called phosphatase-transcriptional activators but without intrinsic DNA-binding activity. However, the spectrum of the Eya1-centered networks is underexplored. Here, we combined transcriptomic, genomic and proteomic approaches to characterize gene regulation by Eya1 in the NPCs. We identified Eya1 target genes, associated cis-regulatory elements and partner proteins. Eya1 preferentially occupies promoter sequences and interacts with general transcription factors (TFs), RNA polymerases, different types of TFs, chromatin-remodeling factors with ATPase or helicase activity, and DNA replication/repair proteins. Intriguingly, we identified REST-binding motifs in 76% of Eya1-occupied sites without H3K27ac-deposition, which were present in many Eya1 target genes upregulated in Eya1-deficient NPCs. Eya1 copurified REST-interacting chromatin-remodeling factors, histone deacetylase/lysine demethylase, and corepressors. Coimmunoprecipitation validated physical interaction between Eya1 and Rest/Hdac1/Cdyl/Hltf in the kidneys. Collectively, our results suggest that through interactions with chromatin-remodeling factors and specialized DNA-binding proteins, Eya1 may modify chromatin structure to facilitate the assembly of regulatory complexes that regulate transcription positively or negatively. These findings provide a mechanistic basis for how Eya1 exerts its activity by forming unique multiprotein complexes in various biological processes to maintain the cellular state of NPCs.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nefronas/citología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Animales , Cromatina/genética , Proteínas Co-Represoras , Proteínas de Unión al ADN/genética , Histona Desacetilasas/metabolismo , Ratones , Complejos Multiproteicos/genética , Nefronas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Proteómica , Factores Generales de Transcripción/genética
6.
Am J Physiol Renal Physiol ; 322(2): F121-F137, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894726

RESUMEN

Normal pregnancy is characterized by massive increases in plasma volume and electrolyte retention. Given that the kidneys regulate homeostasis of electrolytes and volume, the organ undergoes major adaptations in morphology, hemodynamics, and transport to achieve the volume and electrolyte retention required in pregnancy. These adaptations are complex, sometimes counterintuitive, and not fully understood. In addition, the demands of the developing fetus and placenta change throughout pregnancy. For example, during late pregnancy, K+ retention and thus enhanced renal K+ reabsorption are required despite many kaliuretic factors. The goal of this study was to unravel how known adaptive changes along the nephrons contribute to the ability of the kidney to meet volume and electrolyte requirements in mid and late pregnancy. We developed computational models of solute and water transport in the superficial nephron of the kidney of a rat in mid and late pregnancy. The midpregnant and late-pregnant rat superficial nephron models predicted that morphological adaptations and increased activity of Na+/H+ exchanger 3 (NHE3) and epithelial Na+ channel are essential for the enhanced Na+ reabsorption observed during pregnancy. Model simulations showed that for sufficient K+ reabsorption, increased activity of H+-K+-ATPase and decreased K+ secretion along the distal segments is required in both mid and late pregnancy. The model results also suggested that certain known sex differences in renal transporter pattern (e.g., the higher NHE3 protein abundance but lower activity in the proximal tubules of virgin female rats compared with male rats) may serve to better prepare females for the increased transport demand in pregnancy.NEW & NOTEWORTHY Normal pregnancy in mammals is generally characterized by massive changes in plasma volume and electrolyte retention. This study provides insights into how the volume and electrolyte requirement in different pregnancy stages are met by coordinated adaptive changes in the kidney. The model results also suggested that certain known sex differences in the renal transporter pattern may serve to better prepare females for the increased transport demand in pregnancy.


Asunto(s)
Células Epiteliales/metabolismo , Tasa de Filtración Glomerular , Modelos Biológicos , Nefronas/metabolismo , Potasio/metabolismo , Reabsorción Renal , Sodio/metabolismo , Equilibrio Hidroelectrolítico , Adaptación Fisiológica , Animales , Acuaporinas/metabolismo , Canales Epiteliales de Sodio/metabolismo , Femenino , Masculino , Nefronas/citología , Volumen Plasmático , Embarazo , Ratas , Factores Sexuales , Intercambiador 3 de Sodio-Hidrógeno/metabolismo
7.
Development ; 146(8)2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31036548

RESUMEN

Zebrafish kidneys use resident kidney stem cells to replace damaged tubules with new nephrons: the filtration units of the kidney. What stimulates kidney progenitor cells to form new nephrons is not known. Here, we show that wnt9a and wnt9b are induced in the injured kidney at sites where frizzled9b- and lef1-expressing progenitor cells form new nephrons. New nephron aggregates are patterned by Wnt signaling, with high canonical Wnt-signaling cells forming a single cell thick rosette that demarcates: domains of cell proliferation in the elongating nephron; and tubule fusion where the new nephron plumbs into the distal tubule and establishes blood filtrate drainage. Pharmacological blockade of canonical Wnt signaling inhibited new nephron formation after injury by inhibiting cell proliferation, and resulted in loss of polarized rosette structures in the aggregates. Mutation in frizzled9b reduced total kidney nephron number, caused defects in tubule morphology and reduced regeneration of new nephrons after injury. Our results demonstrate an essential role for Wnt/frizzled signaling in adult zebrafish kidney development and regeneration, highlighting conserved mechanisms underlying both mammalian kidney development and kidney stem cell-directed neonephrogenesis in zebrafish.


Asunto(s)
Riñón/citología , Riñón/metabolismo , Nefronas/citología , Nefronas/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regeneración/fisiología , Vía de Señalización Wnt/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
J Am Soc Nephrol ; 32(11): 2815-2833, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34716243

RESUMEN

BACKGROUND: Eya1 is a critical regulator of nephron progenitor cell specification and interacts with Six2 to promote NPC self-renewal. Haploinsufficiency of these genes causes kidney hypoplasia. However, how the Eya1-centered network operates remains unknown. METHODS: We engineered a 2×HA-3×Flag-Eya1 knock-in mouse line and performed coimmunoprecipitation with anti-HA or -Flag to precipitate the multitagged-Eya1 and its associated proteins. Loss-of-function, transcriptome profiling, and genome-wide binding analyses for Eya1's interacting chromatin-remodeling ATPase Brg1 were carried out. We assayed the activity of the cis-regulatory elements co-occupied by Brg1/Six2 in vivo. RESULTS: Eya1 and Six2 interact with the Brg1-based SWI/SNF complex during kidney development. Knockout of Brg1 results in failure of metanephric mesenchyme formation and depletion of nephron progenitors, which has been linked to loss of Eya1 expression. Transcriptional profiling shows conspicuous downregulation of important regulators for nephrogenesis in Brg1-deficient cells, including Lin28, Pbx1, and Dchs1-Fat4 signaling, but upregulation of podocyte lineage, oncogenic, and cell death-inducing genes, many of which Brg1 targets. Genome-wide binding analysis identifies Brg1 occupancy to a distal enhancer of Eya1 that drives nephron progenitor-specific expression. We demonstrate that Brg1 enrichment to two distal intronic enhancers of Pbx1 and a proximal promoter region of Mycn requires Six2 activity and that these Brg1/Six2-bound enhancers govern nephron progenitor-specific expression in response to Six2 activity. CONCLUSIONS: Our results reveal an essential role for Brg1, its downstream pathways, and its interaction with Eya1-Six2 in mediating the fine balance among the self-renewal, differentiation, and survival of nephron progenitors.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/fisiología , Elementos de Facilitación Genéticos , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nefronas/citología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Diferenciación Celular , Autorrenovación de las Células , Inmunoprecipitación de Cromatina , Técnicas de Sustitución del Gen , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Riñón/embriología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Complejos Multiproteicos , Proteínas Nucleares/genética , Mapeo de Interacción de Proteínas , Proteínas Tirosina Fosfatasas/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Transcriptoma
9.
Semin Cell Dev Biol ; 91: 153-168, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30184476

RESUMEN

Decades of research into the molecular and cellular regulation of kidney morphogenesis in rodent models, particularly the mouse, has provided both an atlas of the mammalian kidney and a roadmap for recreating kidney cell types with potential applications for the treatment of kidney disease. With advances in both our capacity to maintain nephron progenitors in culture, reprogram to kidney cell types and direct the differentiation of human pluripotent stem cells to kidney endpoints, renal regeneration via cellular therapy or tissue engineering may be possible. Human kidney models also have potential for disease modelling and drug screening. Such applications will rely upon the accuracy of the model at the cellular level and the capacity for stem-cell derived kidney tissue to recapitulate both normal and diseased kidney tissue. In this review, we will discuss the available cell sources, how well they model the human kidney and how far we are from application either as models or for tissue engineering.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Riñón/fisiología , Nefronas/fisiología , Regeneración , Insuficiencia Renal Crónica/terapia , Animales , Diferenciación Celular , Humanos , Riñón/citología , Nefronas/citología , Células Madre Pluripotentes/citología , Insuficiencia Renal Crónica/fisiopatología , Ingeniería de Tejidos/métodos
10.
Semin Cell Dev Biol ; 91: 147-152, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31178004

RESUMEN

Chronic kidney disease can be understood as a pathological reduction in the number of functional glomeruli. It is a frequent medical problem and one of the major independent risk factors for cardiovascular morbidity and mortality. In humans, glomeruli/nephrons are generated during the prenatal period (glomerular endowment), which may be impaired by multiple conditions. After birth, glomeruli are progressively lost - mostly due to glomerular scarring (focal segmental glomerulosclerosis; FSGS). Multiple independent studies have shown that significant loss of glomerular visceral epithelial cells (podocytes) is sufficient to induce FSGS. It is generally believed that podocytes cannot renew themselves and it has been generally assumed that their number is determined at birth (podocyte endowment). However, there are several lines of experimental evidence showing that podocytes can be replenished in the postnatal period. First, a limited reserve of podocytes has been reported on Bowman's capsule, which may be associated with body growth and increases in glomerular size between childhood and adulthood. Second, two intrinsic progenitor cell niches have been proposed to replenish podocytes throughout adult life and in association with glomerular injury and podocyte loss: parietal epithelial cells and/or cells of the renin lineage. While there is increasing evidence supporting postnatal podocyte gain, controversy remains about the involved signalling pathways and the efficiency of these sources to prevent nephron loss.


Asunto(s)
Células Epiteliales/citología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Glomérulos Renales/citología , Nefronas/citología , Podocitos/citología , Animales , Modelos Animales de Enfermedad , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Glomérulos Renales/crecimiento & desarrollo , Nefronas/crecimiento & desarrollo , Nicho de Células Madre , Células Madre/citología
11.
Semin Cell Dev Biol ; 91: 119-131, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-29857053

RESUMEN

There is a global epidemic of chronic kidney disease (CKD) characterized by a progressive loss of nephrons, ascribed in large part to a rising incidence of hypertension, metabolic syndrome, and type 2 diabetes mellitus. There is a ten-fold variation in nephron number at birth in the general population, and a 50% overall decrease in nephron number in the last decades of life. The vicious cycle of nephron loss stimulating hypertrophy by remaining nephrons and resulting in glomerulosclerosis has been regarded as maladaptive, and only partially responsive to angiotensin inhibition. Advances over the past century in kidney physiology, genetics, and development have elucidated many aspects of nephron formation, structure and function. Parallel advances have been achieved in evolutionary biology, with the emergence of evolutionary medicine, a discipline that promises to provide new insight into the treatment of chronic disease. This review provides a framework for understanding the origins of contemporary developmental nephrology, and recent progress in evolutionary biology. The establishment of evolutionary developmental biology (evo-devo), ecological developmental biology (eco-devo), and developmental origins of health and disease (DOHaD) followed the discovery of the hox gene family, the recognition of the contribution of cumulative environmental stressors to the changing phenotype over the life cycle, and mechanisms of epigenetic regulation. The maturation of evolutionary medicine has contributed to new investigative approaches to cardiovascular disease, cancer, and infectious disease, and promises the same for CKD. By incorporating these principles, developmental nephrology is ideally positioned to answer important questions regarding the fate of nephrons from embryo through senescence.


Asunto(s)
Biología Evolutiva/métodos , Evolución Molecular , Nefronas/metabolismo , Insuficiencia Renal Crónica/genética , Animales , Biología Evolutiva/tendencias , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Nefronas/citología , Nefronas/embriología , Organogénesis/genética , Insuficiencia Renal Crónica/embriología , Insuficiencia Renal Crónica/patología
12.
Semin Cell Dev Biol ; 91: 111-118, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30172047

RESUMEN

Developmental changes in cell fate are tightly regulated by cell-type specific transcription factors. Chromatin reorganization during organismal development ensures dynamic access of developmental regulators to their cognate DNA sequences. Thus, understanding the epigenomic states of promoters and enhancers is of key importance. Recent years have witnessed significant advances in our knowledge of the transcriptional mechanisms of kidney development. Emerging evidence suggests that histone deacetylation by class I HDACs and H3 methylation on lysines 4, 27 and 79 play important roles in regulation of early and late gene expression in the developing kidney. Equally exciting is the realization that nephrogenesis genes in mesenchymal nephron progenitors harbor bivalent chromatin domains which resolve upon differentiation implicating chromatin bivalency in developmental control of gene expression. Here, we review current knowledge of the epigenomic states of nephric cells and current techniques used to study the dynamic chromatin states. These technological advances will provide an unprecedented view of the enhancer landscape during cell fate commitment and help in defining the complex transcriptional networks governing kidney development and disease.


Asunto(s)
Epigénesis Genética , Epigenómica/métodos , Riñón/metabolismo , Nefronas/metabolismo , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/citología , Riñón/embriología , Enfermedades Renales/embriología , Enfermedades Renales/genética , Enfermedades Renales/patología , Nefronas/citología , Nefronas/embriología , Organogénesis/genética
13.
Semin Cell Dev Biol ; 91: 94-103, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30030141

RESUMEN

The filtering component of the kidney, the nephron, arises from a single progenitor population. These nephron progenitor cells (NPCs) both self-renew and differentiate throughout the course of kidney development ensuring sufficient nephron endowment. An appropriate balance of these processes must be struck as deficiencies in nephron numbers are associated with hypertension and kidney disease. This review will discuss the mechanisms and molecules supporting NPC maintenance and differentiation. A focus on recent work will highlight new molecular insights into NPC regulation and their dynamic behavior in both space and time.


Asunto(s)
Riñón/citología , Nefronas/citología , Podocitos/citología , Células Madre/citología , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/embriología , Riñón/metabolismo , Nefronas/metabolismo , Organogénesis/genética , Podocitos/metabolismo , Células Madre/metabolismo
14.
Dev Biol ; 464(2): 176-187, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32504627

RESUMEN

Chromatin-remodeling complexes play critical roles in establishing gene expression patterns in response to developmental signals. How these epigenetic regulators determine the fate of progenitor cells during development of specific organs is not well understood. We found that genetic deletion of Brg1 (Smarca4), the core enzymatic protein in SWI/SNF, in nephron progenitor cells leads to severe renal hypoplasia. Nephron progenitor cells were depleted in Six2-Cre, Brg1flx/flx mice due to reduced cell proliferation. This defect in self-renewal, together with impaired differentiation resulted in a profound nephron deficit in Brg1 mutant kidneys. Sall1, a transcription factor that is required for expansion and maintenance of nephron progenitors, associates with SWI/SNF. Brg1 and Sall1 bind promoters of many progenitor cell genes and regulate expression of key targets that promote their proliferation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , ADN Helicasas/metabolismo , Nefronas/embriología , Proteínas Nucleares/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Células COS , Chlorocebus aethiops , ADN Helicasas/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Nefronas/citología , Proteínas Nucleares/genética , Células Madre/citología , Factores de Transcripción/genética
15.
J Biol Chem ; 295(33): 11542-11558, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32554463

RESUMEN

SIX2 (SIX homeobox 2)-positive nephron progenitor cells (NPCs) give rise to all epithelial cell types of the nephron, the filtering unit of the kidney. NPCs have a limited lifespan and are depleted near the time of birth. Epigenetic factors are implicated in the maintenance of organ-restricted progenitors such as NPCs, but the chromatin-based mechanisms are incompletely understood. Here, using a combination of gene targeting, chromatin profiling, and single-cell RNA analysis, we examined the role of the murine histone 3 Lys-27 (H3K27) methyltransferases EZH1 (enhancer of zeste 1) and EZH2 in NPC maintenance. We found that EZH2 expression correlates with NPC growth potential and that EZH2 is the dominant H3K27 methyltransferase in NPCs and epithelial descendants. Surprisingly, NPCs lacking H3K27 trimethylation maintained their progenitor state but cycled slowly, leading to a smaller NPC pool and formation of fewer nephrons. Unlike Ezh2 loss of function, dual inactivation of Ezh1 and Ezh2 triggered overexpression of the transcriptional repressor Hes-related family BHLH transcription factor with YRPW motif 1 (Hey1), down-regulation of Six2, and unscheduled activation of Wnt4-driven differentiation, resulting in early termination of nephrogenesis and severe renal dysgenesis. Double-mutant NPCs also overexpressed the SIX family member Six1 However, in this context, SIX1 failed to maintain NPC stemness. At the chromatin level, EZH1 and EZH2 restricted accessibility to AP-1-binding motifs, and their absence promoted a regulatory landscape akin to differentiated and nonlineage cells. We conclude that EZH2 is required for NPC renewal potential and that tempering of the differentiation program requires cooperation of both EZH1 and EZH2.


Asunto(s)
Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Nefronas/citología , Complejo Represivo Polycomb 2/metabolismo , Células Madre/citología , Animales , Supervivencia Celular , Células Cultivadas , Ratones , Nefronas/metabolismo , Células Madre/metabolismo
16.
Biochem Biophys Res Commun ; 558: 231-238, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32113685

RESUMEN

Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.


Asunto(s)
Proteína Morfogenética Ósea 7/metabolismo , Inhibidores de las Cinasas Janus/farmacología , Nefronas/citología , Nefronas/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 7/administración & dosificación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Medios de Cultivo , Evaluación Preclínica de Medicamentos , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Janus Quinasa 3/antagonistas & inhibidores , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Nefronas/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas
17.
Development ; 145(14)2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29945864

RESUMEN

Epigenetic regulation of gene expression has a crucial role allowing for the self-renewal and differentiation of stem and progenitor populations during organogenesis. The mammalian kidney maintains a population of self-renewing stem cells that differentiate to give rise to thousands of nephrons, which are the functional units that carry out filtration to maintain physiological homeostasis. The polycomb repressive complex 2 (PRC2) epigenetically represses gene expression during development by placing the H3K27me3 mark on histone H3 at promoter and enhancer sites, resulting in gene silencing. To understand the role of PRC2 in nephron differentiation, we conditionally inactivated the Eed gene, which encodes a nonredundant component of the PRC2 complex, in nephron progenitor cells. Resultant kidneys were smaller and showed premature loss of progenitor cells. The progenitors in Eed mutant mice that were induced to differentiate did not develop into properly formed nephrons. Lhx1, normally expressed in the renal vesicle, was overexpressed in kidneys of Eed mutant mice. Thus, PRC2 has a crucial role in suppressing the expression of genes that maintain the progenitor state, allowing nephron differentiation to proceed.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Nefronas/embriología , Complejo Represivo Polycomb 2/biosíntesis , Células Madre/metabolismo , Animales , Proteínas con Homeodominio LIM/biosíntesis , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Transgénicos , Mutación , Nefronas/citología , Complejo Represivo Polycomb 2/genética , Células Madre/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
18.
Development ; 145(13)2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945868

RESUMEN

Normal kidney function depends on the proper development of the nephron: the functional unit of the kidney. Reciprocal signaling interactions between the stroma and nephron progenitor compartment have been proposed to control nephron development. Here, we show that removal of hedgehog intracellular effector smoothened (Smo-deficient mutants) in the cortical stroma results in an abnormal renal capsule, and an expanded nephron progenitor domain with an accompanying decrease in nephron number via a block in epithelialization. We show that stromal-hedgehog-Smo signaling acts through a GLI3 repressor. Whole-kidney RNA sequencing and analysis of FACS-isolated stromal cells identified impaired TGFß2 signaling in Smo-deficient mutants. We show that neutralization and knockdown of TGFß2 in explants inhibited nephrogenesis. In addition, we demonstrate that concurrent deletion of Tgfbr2 in stromal and nephrogenic cells in vivo results in decreased nephron formation and an expanded nephrogenic precursor domain similar to that observed in Smo-deficient mutant mice. Together, our data suggest a mechanism whereby a stromal hedgehog-TGFß2 signaling axis acts to control nephrogenesis.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas Hedgehog/metabolismo , Nefronas/embriología , Transducción de Señal/fisiología , Receptor Smoothened/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/genética , Ratones , Ratones Noqueados , Nefronas/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptor Smoothened/genética , Células del Estroma/citología , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta2/genética , Proteína Gli3 con Dedos de Zinc/genética , Proteína Gli3 con Dedos de Zinc/metabolismo
19.
Development ; 145(10)2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29712641

RESUMEN

Nephron progenitor cells (NPCs) are Six2-positive metanephric mesenchyme cells, which undergo self-renewal and differentiation to give rise to nephrons until the end of nephrogenesis. Histone deacetylases (HDACs) are a group of epigenetic regulators that control cell fate, but their role in balancing NPC renewal and differentiation is unknown. Here, we report that NPC-specific deletion of Hdac1 and Hdac2 genes in mice results in early postnatal lethality owing to renal hypodysplasia and loss of NPCs. HDAC1/2 interact with the NPC renewal regulators Six2, Osr1 and Sall1, and are co-bound along with Six2 on the Six2 enhancer. Although the mutant NPCs differentiate into renal vesicles (RVs), Hdac1/2 mutant kidneys lack nascent nephrons or mature glomeruli, a phenocopy of Lhx1 mutants. Transcriptional profiling and network analysis identified disrupted expression of Lhx1 and its downstream genes, Dll1 and Hnf1a/4a, as key mediators of the renal phenotype. Finally, although HDAC1/2-deficient NPCs and RVs overexpress hyperacetylated p53, Trp53 deletion failed to rescue the renal dysgenesis. We conclude that the epigenetic regulators HDAC1 and HDAC2 control nephrogenesis via interactions with the transcriptional programs of nephron progenitors and renal vesicles.


Asunto(s)
Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Nefronas/embriología , Organogénesis/genética , Células Madre/citología , Transcripción Genética/genética , Animales , Proteínas de Unión al Calcio , Línea Celular , Proliferación Celular/genética , Células HEK293 , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Factor Nuclear 4 del Hepatocito/biosíntesis , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Enfermedades Renales/genética , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Noqueados , Nefronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/genética
20.
FASEB J ; 34(4): 5782-5799, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32141129

RESUMEN

Low nephron number results in an increased risk of developing hypertension and chronic kidney disease. Intrauterine growth restriction is associated with a nephron deficit in humans, and is commonly caused by placental insufficiency, which results in fetal hypoxia. The underlying mechanisms by which hypoxia impacts kidney development are poorly understood. microRNA-210 is the most consistently induced microRNA in hypoxia and is known to promote cell survival in a hypoxic environment. In this study, the role of microRNA-210 in kidney development was evaluated using a global microRNA-210 knockout mouse. A male-specific 35% nephron deficit in microRNA-210 knockout mice was observed. Wnt/ß-catenin signaling, a pathway crucial for nephron differentiation, was misregulated in male kidneys with increased expression of the canonical Wnt target lymphoid enhancer binding factor 1. This coincided with increased expression of caspase-8-associated protein 2, a known microRNA-210 target and apoptosis signal transducer. Together, these data are consistent with a sex-specific requirement for microRNA-210 in kidney development.


Asunto(s)
Diferenciación Celular , Hipoxia/fisiopatología , MicroARNs/genética , Nefronas/citología , Organogénesis , Animales , Apoptosis , Femenino , Masculino , Ratones , Ratones Noqueados , Nefronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA