Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 432
Filtrar
1.
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
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.
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
4.
Cell Syst ; 15(7): 649-661.e9, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38981488

RESUMEN

Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.


Asunto(s)
Organoides , Organoides/citología , Organoides/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Nefronas/citología , Diferenciación Celular/fisiología , Células Madre/citología
5.
Cell Stem Cell ; 31(6): 921-939.e17, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38692273

RESUMEN

Nephron progenitor cells (NPCs) self-renew and differentiate into nephrons, the functional units of the kidney. Here, manipulation of p38 and YAP activity allowed for long-term clonal expansion of primary mouse and human NPCs and induced NPCs (iNPCs) from human pluripotent stem cells (hPSCs). Molecular analyses demonstrated that cultured iNPCs closely resemble primary human NPCs. iNPCs generated nephron organoids with minimal off-target cell types and enhanced maturation of podocytes relative to published human kidney organoid protocols. Surprisingly, the NPC culture medium uncovered plasticity in human podocyte programs, enabling podocyte reprogramming to an NPC-like state. Scalability and ease of genome editing facilitated genome-wide CRISPR screening in NPC culture, uncovering genes associated with kidney development and disease. Further, NPC-directed modeling of autosomal-dominant polycystic kidney disease (ADPKD) identified a small-molecule inhibitor of cystogenesis. These findings highlight a broad application for the reported iNPC platform in the study of kidney development, disease, plasticity, and regeneration.


Asunto(s)
Nefronas , Organoides , Animales , Organoides/citología , Organoides/metabolismo , Humanos , Nefronas/citología , Ratones , Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Podocitos/metabolismo , Podocitos/citología , Riñón/patología , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Modelos Biológicos , Edición Génica
6.
Matrix Biol ; 131: 30-45, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38788809

RESUMEN

Renal development is a complex process in which two major processes, tubular branching and nephron development, regulate each other reciprocally. Our previous findings have indicated that collagen XVIII (ColXVIII), an extracellular matrix protein, affects the renal branching morphogenesis. We investigate here the role of ColXVIII in nephron formation and the behavior of nephron progenitor cells (NPCs) using isoform-specific ColXVIII knockout mice. The results show that the short ColXVIII isoform predominates in the early epithelialized nephron structures whereas the two longer isoforms are expressed only in the later phases of glomerular formation. Meanwhile, electron microscopy showed that the ColXVIII mutant embryonic kidneys have ultrastructural defects at least from embryonic day 16.5 onwards. Similar structural defects had previously been observed in adult ColXVIII-deficient mice, indicating a congenital origin. The lack of ColXVIII led to a reduced NPC population in which changes in NPC proliferation and maintenance and in macrophage influx were perceived to play a role. The changes in NPC behavior in turn led to notably reduced overall nephron formation. In conclusion, the results show that ColXVIII has multiple roles in renal development, both in ureteric branching and in NPC behavior.


Asunto(s)
Matriz Extracelular , Ratones Noqueados , Nefronas , Células Madre , Animales , Nefronas/metabolismo , Nefronas/citología , Nefronas/crecimiento & desarrollo , Ratones , Matriz Extracelular/metabolismo , Células Madre/metabolismo , Células Madre/citología , Proliferación Celular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Colágeno/metabolismo , Colágeno/genética
7.
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
8.
Cell Rep ; 39(11): 110933, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705028

RESUMEN

Generation of new kidneys can be useful in various research fields, including organ transplantation. However, generating renal stroma, an important component tissue for structural support, endocrine function, and kidney development, remains difficult. Organ generation using an animal developmental niche can provide an appropriate in vivo environment for renal stroma differentiation. Here, we generate rat renal stroma with endocrine capacity by removing mouse stromal progenitor cells (SPCs) from the host developmental niche and transplanting rat SPCs. Furthermore, we develop a method to replace both nephron progenitor cells (NPCs) and SPCs, called the interspecies dual replacement of the progenitor (i-DROP) system, and successfully generate functional chimeric kidneys containing rat nephrons and stroma. This method can generate renal tissue from progenitors and reduce xenotransplant rejection. Moreover, it is a safe method, as donor cells do not stray into nontarget organs, thus accelerating research on stem cells, chimeras, and xenotransplantation.


Asunto(s)
Riñón , Nefronas , Nicho de Células Madre , Células Madre , Animales , Diferenciación Celular , Quimera , Riñón/citología , Ratones , Nefronas/citología , Ratas , Células Madre/citología
9.
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
10.
Nat Commun ; 12(1): 6332, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732708

RESUMEN

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


Asunto(s)
Organogénesis/fisiología , Percepción/fisiología , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Diferenciación Celular , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio , Riñón/citología , Riñón/patología , Masculino , Proteínas de la Membrana , Ratones , Nefronas/citología , Proteínas del Tejido Nervioso , Nicho de Células Madre , Células Madre/citología , Factores de Transcripción/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Vía de Señalización Wnt
11.
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
12.
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
13.
Sci Rep ; 11(1): 9123, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33907292

RESUMEN

PAX2 is a transcription factor essential for kidney development and the main causative gene for renal coloboma syndrome (RCS). The mechanisms of PAX2 action during kidney development have been evaluated in mice but not in humans. This is a critical gap in knowledge since important differences have been reported in kidney development in the two species. In the present study, we hypothesized that key human PAX2-dependent kidney development genes are differentially expressed in nephron progenitor cells from induced pluripotent stem cells (iPSCs) in patients with RCS relative to healthy individuals. Cap analysis of gene expression revealed 189 candidate promoters and 71 candidate enhancers that were differentially activated by PAX2 in this system in three patients with RCS with PAX2 mutations. By comparing this list with the list of candidate Pax2-regulated mouse kidney development genes obtained from the Functional Annotation of the Mouse/Mammalian (FANTOM) database, we prioritized 17 genes. Furthermore, we ranked three genes-PBX1, POSTN, and ITGA9-as the top candidates based on closely aligned expression kinetics with PAX2 in the iPSC culture system and susceptibility to suppression by a Pax2 inhibitor in cultured mouse embryonic kidney explants. Identification of these genes may provide important information to clarify the pathogenesis of RCS, human kidney development, and kidney regeneration.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/crecimiento & desarrollo , Factor de Transcripción PAX2/genética , Adulto , Animales , Moléculas de Adhesión Celular/genética , Linaje de la Célula , Coloboma/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas , Integrinas/genética , Riñón/citología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Nefronas/citología , Nefronas/fisiología , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Insuficiencia Renal/patología
14.
Nat Commun ; 12(1): 2277, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859189

RESUMEN

Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene-regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Nefronas/crecimiento & desarrollo , Organogénesis/genética , Insuficiencia Renal Crónica/genética , Animales , Comunicación Celular , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Epigenómica , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Ratones , Nefronas/citología , Podocitos/fisiología , Polimorfismo de Nucleótido Simple , RNA-Seq , Insuficiencia Renal Crónica/patología , Análisis de la Célula Individual , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
15.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669059

RESUMEN

The renin-angiotensin-aldosterone system (RAAS) is implicated in hypertension and kidney disease. The developing kidney can be programmed by various early-life insults by so-called renal programming, resulting in hypertension and kidney disease in adulthood. This theory is known as developmental origins of health and disease (DOHaD). Conversely, early RAAS-based interventions could reverse program processes to prevent a disease from occurring by so-called reprogramming. In the current review, we mainly summarize (1) the current knowledge on the RAAS implicated in renal programming; (2) current evidence supporting the connections between the aberrant RAAS and other mechanisms behind renal programming, such as oxidative stress, nitric oxide deficiency, epigenetic regulation, and gut microbiota dysbiosis; and (3) an overview of how RAAS-based reprogramming interventions may prevent hypertension and kidney disease of developmental origins. To accelerate the transition of RAAS-based interventions for prevention of hypertension and kidney disease, an extended comprehension of the RAAS implicated in renal programming is needed, as well as a greater focus on further clinical translation.


Asunto(s)
Hipertensión/metabolismo , Enfermedades Renales/metabolismo , Riñón/crecimiento & desarrollo , Nefronas/crecimiento & desarrollo , Sistema Renina-Angiotensina , Renina/metabolismo , Adulto , Animales , Modelos Animales de Enfermedad , Disbiosis/metabolismo , Epigénesis Genética , Humanos , Hipertensión/genética , Riñón/metabolismo , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Nefronas/citología , Nefronas/metabolismo , Óxido Nítrico/deficiencia , Óxido Nítrico/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiología
16.
Elife ; 102021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33587034

RESUMEN

The canonical Wnt pathway transcriptional co-activator ß-catenin regulates self-renewal and differentiation of mammalian nephron progenitor cells (NPCs). We modulated ß-catenin levels in NPC cultures using the GSK3 inhibitor CHIR99021 (CHIR) to examine opposing developmental actions of ß-catenin. Low CHIR-mediated maintenance and expansion of NPCs are independent of direct engagement of TCF/LEF/ß-catenin transcriptional complexes at low CHIR-dependent cell-cycle targets. In contrast, in high CHIR, TCF7/LEF1/ß-catenin complexes replaced TCF7L1/TCF7L2 binding on enhancers of differentiation-promoting target genes. Chromosome confirmation studies showed pre-established promoter-enhancer connections to these target genes in NPCs. High CHIR-associated de novo looping was observed in positive transcriptional feedback regulation to the canonical Wnt pathway. Thus, ß-catenin's direct transcriptional role is restricted to the induction of NPCs, where rising ß-catenin levels switch inhibitory TCF7L1/TCF7L2 complexes to activating LEF1/TCF7 complexes at primed gene targets poised for rapid initiation of a nephrogenic program.


Asunto(s)
Factor de Unión 1 al Potenciador Linfoide/metabolismo , Nefronas/metabolismo , Células Madre/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Nefronas/citología , Nefronas/embriología , Regiones Promotoras Genéticas , Unión Proteica , Células Madre/citología , Proteína 1 Similar al Factor de Transcripción 7/genética , Factores de Transcripción/genética
17.
Sci Rep ; 11(1): 73, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420268

RESUMEN

Kidney development requires the coordinated growth and differentiation of multiple cells. Despite recent single cell profiles in nephrogenesis research, tools for data analysis are rapidly developing, and offer an opportunity to gain additional insight into kidney development. In this study, single-cell RNA sequencing data obtained from embryonic mouse kidney were re-analyzed. Manifold learning based on partition-based graph-abstraction coordinated cells, reflecting their expected lineage relationships. Consequently, the coordination in combination with ForceAtlas2 enabled the inference of parietal epithelial cells of Bowman's capsule and the inference of cells involved in the developmental process from the S-shaped body to each nephron segment. RNA velocity suggested developmental sequences of proximal tubules and podocytes. In combination with a Markov chain algorithm, RNA velocity suggested the self-renewal processes of nephron progenitors. NicheNet analyses suggested that not only cells belonging to ureteric bud and stroma, but also endothelial cells, macrophages, and pericytes may contribute to the differentiation of cells from nephron progenitors. Organ culture of embryonic mouse kidney demonstrated that nerve growth factor, one of the nephrogenesis-related factors inferred by NicheNet, contributed to mitochondrial biogenesis in developing distal tubules. These approaches suggested previously unrecognized aspects of the underlying mechanisms for kidney development.


Asunto(s)
Comunicación Celular , Riñón/embriología , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Animales , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica/genética , Riñón/citología , Ratones , Ratones Endogámicos C57BL , Nefronas/citología , Nefronas/embriología , Análisis de Secuencia de ARN/métodos
18.
J Dev Orig Health Dis ; 12(2): 179-183, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-31983353

RESUMEN

The mammalian kidney is a complex organ, requiring the concerted function of up to millions of nephrons. The number of nephrons is constant after nephrogenesis during development, and nephron loss over a life span can lead to susceptibility to acute or chronic kidney disease. New technologies are under development to count individual nephrons in the kidney in vivo. This review outlines these technologies and highlights their relevance to studies of human renal development and disease.


Asunto(s)
Investigación Biomédica/tendencias , Diagnóstico por Imagen/métodos , Enfermedades Renales/patología , Nefronas/citología , Organogénesis , Animales , Humanos , Enfermedades Renales/diagnóstico por imagen , Nefronas/diagnóstico por imagen
19.
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
20.
Cell Rep ; 32(11): 108130, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937125

RESUMEN

Animal fetuses may be used for the regeneration of human organs. We have previously generated a transgenic mouse model that allows diphtheria toxin (DT)-induced ablation of Six2-positive nephron progenitor cells (NPCs). Elimination of existing native host NPCs enables their replacement with donor NPCs, which can generate neo-nephrons. However, this system cannot be applied to human NPCs, because DT induces apoptosis in human cells. Therefore, the present study presents a transgenic mouse model for the ablation of NPCs using tamoxifen, which does not affect human cells. Using this system, we successfully regenerate interspecies neo-nephrons, which exhibit urine-producing abilities, from transplanted rat NPCs in a mouse host. Transplantation of human induced pluripotent stem cell (iPSC)-derived NPCs results in differentiation into renal vesicles, which connect to the ureteric bud of the host. Thus, we demonstrate the possibility of the regeneration of human kidneys derived from human iPSC-derived NPCs via NPC replacement.


Asunto(s)
Nefronas/citología , Regeneración , Células Madre/citología , Animales , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones Endogámicos C57BL , Nefronas/efectos de los fármacos , Nefronas/ultraestructura , Especificidad de Órganos , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Especificidad de la Especie , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo , Vejiga Urinaria/embriología , Micción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA