Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34948246

RESUMO

BACKGROUND: Regenerative medicine strategies employing nephron progenitor cells (NPCs) are a viable approach that is worthy of substantial consideration as a promising cell source for kidney diseases. However, the generation of induced nephron progenitor-like cells (iNPCs) from human somatic cells remains a major challenge. Here, we describe a novel method for generating NPCs from human urine-derived cells (UCs) that can undergo long-term expansion in a serum-free condition. RESULTS: Here, we generated iNPCs from human urine-derived cells by forced expression of the transcription factors OCT4, SOX2, KLF4, c-MYC, and SLUG, followed by exposure to a cocktail of defined small molecules. These iNPCs resembled human embryonic stem cell-derived NPCs in terms of their morphology, biological characteristics, differentiation potential, and global gene expression and underwent a long-term expansion in serum-free conditions. CONCLUSION: This study demonstrates that human iNPCs can be readily generated and expanded, which will facilitate their broad applicability in a rapid, efficient, and patient-specific manner, particularly holding the potential as a transplantable cell source for patients with kidney disease.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Néfrons/metabolismo , Diferenciação Celular/genética , Reprogramação Celular/genética , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Néfrons/crescimento & desenvolvimento , Néfrons/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Urina/citologia
2.
Physiol Genomics ; 53(12): 509-517, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34704838

RESUMO

Preterm neonates are at a high risk for nephron loss under adverse clinical conditions. Renal damage potentially collides with postnatal nephrogenesis. Recent animal studies suggest that nephron loss within this vulnerable phase leads to renal damage later in life. Nephrogenic pathways are commonly reactivated after kidney injury supporting renal regeneration. We hypothesized that nephron loss during nephrogenesis affects renal development, which, in turn, impairs tissue repair after secondary injury. Neonates prior to 36 wk of gestation show an active nephrogenesis. In rats, nephrogenesis is ongoing until day 10 after birth. Mimicking the situation of severe nephron loss during nephrogenesis, male pups were uninephrectomized at day 1 of life (UNXd1). A second group of males was uninephrectomized at postnatal day 14 (UNXd14), after terminated nephrogenesis. Age-matched controls were sham operated. Three days after uninephrectomy transcriptional changes in the right kidney were analyzed by RNA-sequencing, followed by functional pathway analysis. In UNXd1, 1,182 genes were differentially regulated, but only 143 genes showed a regulation both in UNXd1 and UNXd14. The functional groups "renal development" and "kidney injury" were among the most differentially regulated groups and revealed distinctive alterations. Reduced expression of candidate genes concerning renal development (Bmp7, Gdnf, Pdgf-B, Wt1) and injury (nephrin, podocin, Tgf-ß1) were detected. The downregulation of Bmp7 and Gdnf persisted until day 28. In UNXd14, Six2 was upregulated and Pax2 was downregulated. We conclude that nephron loss during nephrogenesis affects renal development and induces a specific regulation of genes that might hinder tissue repair after secondary kidney injury.


Assuntos
Injúria Renal Aguda/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento , Néfrons/crescimento & desenvolvimento , Néfrons/patologia , Organogênese/genética , Regulação para Cima/genética , Animais , Animais Recém-Nascidos/cirurgia , Proteína Morfogenética Óssea 7/genética , Estudos de Casos e Controles , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteínas de Homeodomínio/genética , Masculino , Nefrectomia/métodos , Fator de Transcrição PAX2/genética , RNA-Seq/métodos , Ratos , Ratos Wistar , Transcriptoma/genética
3.
J Am Soc Nephrol ; 32(9): 2255-2272, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34341180

RESUMO

BACKGROUND: Kidney function requires continuous blood filtration by glomerular capillaries. Disruption of glomerular vascular development or maintenance contributes to the pathogenesis of kidney diseases, but the signaling events regulating renal endothelium development remain incompletely understood. Here, we discovered a novel role of Slit2-Robo signaling in glomerular vascularization. Slit2 is a secreted polypeptide that binds to transmembrane Robo receptors and regulates axon guidance as well as ureteric bud branching and angiogenesis. METHODS: We performed Slit2-alkaline phosphatase binding to kidney cryosections from mice with or without tamoxifen-inducible Slit2 or Robo1 and -2 deletions, and we characterized the phenotypes using immunohistochemistry, electron microscopy, and functional intravenous dye perfusion analysis. RESULTS: Only the glomerular endothelium, but no other renal endothelial compartment, responded to Slit2 in the developing kidney vasculature. Induced Slit2 gene deletion or Slit2 ligand trap at birth affected nephrogenesis and inhibited vascularization of developing glomeruli by reducing endothelial proliferation and migration, leading to defective cortical glomerular perfusion and abnormal podocyte differentiation. Global and endothelial-specific Robo deletion showed that both endothelial and epithelial Robo receptors contributed to glomerular vascularization. CONCLUSIONS: Our study provides new insights into the signaling pathways involved in glomerular vascular development and identifies Slit2 as a potential tool to enhance glomerular angiogenesis.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/genética , Glomérulos Renais/irrigação sanguínea , Néfrons/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Animais , Animais Recém-Nascidos , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Camundongos , Néfrons/patologia , Transdução de Sinais , Proteínas Roundabout
4.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34032268

RESUMO

Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Néfrons/embriologia , Néfrons/crescimento & desenvolvimento , Organogênese/genética , Via de Sinalização Wnt/genética , Animais , Proteína Axina/metabolismo , Diferenciação Celular/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/citologia , Proteínas Wnt/metabolismo
5.
Nat Commun ; 12(1): 2277, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859189

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Néfrons/crescimento & desenvolvimento , Organogênese/genética , Insuficiência Renal Crônica/genética , Animais , Comunicação Celular , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Epigenômica , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Loci Gênicos/genética , Estudo de Associação Genômica Ampla , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Néfrons/citologia , Podócitos/fisiologia , Polimorfismo de Nucleotídeo Único , RNA-Seq , Insuficiência Renal Crônica/patologia , Análise de Célula Única , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo
6.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33669059

RESUMO

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.


Assuntos
Hipertensão/metabolismo , Nefropatias/metabolismo , Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Sistema Renina-Angiotensina , Renina/metabolismo , Adulto , Animais , Modelos Animais de Doenças , Disbiose/metabolismo , Epigênese Genética , Humanos , Hipertensão/genética , Rim/metabolismo , Nefropatias/enzimologia , Nefropatias/genética , Néfrons/citologia , Néfrons/metabolismo , Óxido Nítrico/deficiência , Óxido Nítrico/metabolismo , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiologia
7.
STAR Protoc ; 2(1): 100314, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33554147

RESUMO

Renal progenitor cells induced from pluripotent stem cells have attracted attention as a cell source for organ regeneration. Here, we report an in vivo protocol for the regeneration of urine-producing nephrons, i.e., neo-nephrons, in mice. We outline steps to transplant exogenous renal progenitor cells into the nephrogenic zone of transgenic mice and subsequently analyze these neo-nephrons. For complete details on the use and execution of this protocol, please refer to Fujimoto et al. (2020).


Assuntos
Separação Celular/métodos , Néfrons/crescimento & desenvolvimento , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular/fisiologia , Humanos , Rim/citologia , Camundongos , Camundongos Transgênicos , Organogênese/fisiologia , Células-Tronco Pluripotentes/fisiologia , Ratos , Regeneração/fisiologia , Células-Tronco/metabolismo
8.
Sci Rep ; 10(1): 22097, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33328501

RESUMO

In-vivo single cell clonal analysis in the adult mouse kidney has previously shown lineage-restricted clonal proliferation within varying nephron segments as a mechanism responsible for cell replacement and local regeneration. To analyze ex-vivo clonal growth, we now preformed limiting dilution to generate genuine clonal cultures from one single human renal epithelial cell, which can give rise to up to 3.4 * 106 cells, and analyzed their characteristics using transcriptomics. A comparison between clonal cultures revealed restriction to either proximal or distal kidney sub-lineages with distinct cellular and molecular characteristics; rapidly amplifying de-differentiated clones and a stably proliferating cuboidal epithelial-appearing clones, respectively. Furthermore, each showed distinct molecular features including cell-cycle, epithelial-mesenchymal transition, oxidative phosphorylation, BMP signaling pathway and cell surface markers. In addition, analysis of clonal versus bulk cultures show early clones to be more quiescent, with elevated expression of renal developmental genes and overall reduction in renal identity markers, but with an overlapping expression of nephron segment identifiers and multiple identity. Thus, ex-vivo clonal growth mimics the in-vivo situation displaying lineage-restricted precursor characteristics of mature renal cells. These data suggest that for reconstruction of varying renal lineages with human adult kidney based organoid technology and kidney regeneration ex-vivo, use of multiple heterogeneous precursors is warranted.


Assuntos
Evolução Clonal/genética , Rim/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Regeneração/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Biologia Computacional , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/genética , Humanos , Rim/citologia , Mesoderma/metabolismo , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Cultura Primária de Células , Análise de Célula Única , Células-Tronco/citologia
9.
Biomolecules ; 10(10)2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33019532

RESUMO

In mammals, the novel protein fibroblast growth factor receptor-like 1 (FGFRL1) is involved in the development of metanephric kidneys. It appears that this receptor controls a crucial transition of the induced metanephric mesenchyme to epithelial renal vesicles, which further develop into functional nephrons. FGFRL1 knockout mice lack metanephric kidneys and do not express any fibroblast growth factor (FGF) 8 in the metanephric mesenchyme, suggesting that FGFRL1 and FGF8 play a decisive role during kidney formation. FGFRL1 consists of three extracellular immunoglobulin (Ig) domains (Ig1-Ig2-Ig3), a transmembrane domain and a short intracellular domain. We have prepared the extracellular domain (Ig123), the three individual Ig domains (Ig1, Ig2, Ig3) as well as all combinations containing two Ig domains (Ig12, Ig23, Ig13) in recombinant form in human cells. All polypeptides that contain the Ig2 domain (Ig123, Ig12, Ig23, Ig2) were found to interact with FGF8 with very high affinity, whereas all constructs that lack the Ig2 domain (Ig1, Ig3, Ig13) poorly interacted with FGF8 as shown by ELISA and surface plasmon resonance. It is therefore likely that FGFRL1 represents a physiological receptor for FGF8 in the kidney and that the ligand primarily binds to the Ig2 domain of the receptor. With Biacore experiments, we also measured the affinity of FGF8 for the different constructs. All constructs containing the Ig2 domain showed a rapid association and a slow dissociation phase, from which a KD of 2-3 × 10-9 M was calculated. Our data support the hypothesis that binding of FGF8 to FGFRL1 could play an important role in driving the formation of nephrons in the developing kidney.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Domínios de Imunoglobulina/genética , Rim/crescimento & desenvolvimento , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/genética , Animais , Transição Epitelial-Mesenquimal/genética , Humanos , Rim/metabolismo , Ligantes , Camundongos , Camundongos Knockout , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Ressonância de Plasmônio de Superfície
10.
Tissue Barriers ; 8(4): 1832844, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33092489

RESUMO

The kidney is a complex organ that performs essential functions such as blood filtration and fluid homeostasis, among others. Recent years have heralded significant advancements in our knowledge of the mechanisms that control kidney formation. Here, we provide an overview of vertebrate renal development with a focus on nephrogenesis, the process of generating the epithelialized functional units of the kidney. These steps begin with intermediate mesoderm specification and proceed all the way to the terminally differentiated nephron cell, with many detailed stages in between. The establishment of nephron architecture with proper cellular barriers is vital throughout these processes. Continuously striving to gain further insights into nephrogenesis can ultimately lead to a better understanding and potential treatments for developmental maladies such as Congenital Anomalies of the Kidney and Urinary Tract (CAKUT).


Assuntos
Néfrons/crescimento & desenvolvimento , Organogênese/fisiologia , Vertebrados/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Humanos
11.
Development ; 147(23)2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33028614

RESUMO

A functional vertebrate kidney relies on structural units called nephrons, which are epithelial tubules with a sequence of segments each expressing a distinct repertoire of solute transporters. The transcriptiona`l codes driving regional specification, solute transporter program activation and terminal differentiation of segment populations remain poorly understood. Here, we demonstrate that the KCTD15 paralogs kctd15a and kctd15b function in concert to restrict distal early (DE)/thick ascending limb (TAL) segment lineage assignment in the developing zebrafish pronephros by repressing Tfap2a activity. During renal ontogeny, expression of these factors colocalized with tfap2a in distal tubule precursors. kctd15a/b loss primed nephron cells to adopt distal fates by driving slc12a1, kcnj1a.1 and stc1 expression. These phenotypes were the result of Tfap2a hyperactivity, where kctd15a/b-deficient embryos exhibited increased abundance of this transcription factor. Interestingly, tfap2a reciprocally promoted kctd15a and kctd15b transcription, unveiling a circuit of autoregulation operating in nephron progenitors. Concomitant kctd15b knockdown with tfap2a overexpression further expanded the DE population. Our study reveals that a transcription factor-repressor feedback module employs tight regulation of Tfap2a and Kctd15 kinetics to control nephron segment fate choice and differentiation during kidney development.


Assuntos
Desenvolvimento Embrionário/genética , Rim/crescimento & desenvolvimento , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fator de Transcrição AP-2/genética , Proteínas de Peixe-Zebra/genética , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião não Mamífero , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Rim/metabolismo , Túbulos Renais/crescimento & desenvolvimento , Túbulos Renais/metabolismo , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Organogênese/genética , Transdução de Sinais/genética , Membro 1 da Família 12 de Carreador de Soluto/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
12.
J Cell Physiol ; 235(12): 9958-9973, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32474911

RESUMO

Nephron loss stimulates residual functioning nephrons to undergo compensatory growth. Excessive nephron growth may be a maladaptive response that sets the stage for progressive nephron damage, leading to kidney failure. To date, however, the mechanism of nephron growth remains incompletely understood. Our previous study revealed that class III phosphatidylinositol-3-kinase (Pik3c3) is activated in the remaining kidney after unilateral nephrectomy (UNX)-induced nephron loss, but previous studies failed to generate a Pik3c3 gene knockout animal model. Global Pik3c3 deletion results in embryonic lethality. Given that renal proximal tubule cells make up the bulk of the kidney and undergo the most prominent hypertrophic growth after UNX, in this study we used Cre-loxP-based approaches to demonstrate for the first time that tamoxifen-inducible SLC34a1 promoter-driven CreERT2 recombinase-mediated downregulation of Pik3c3 expression in renal proximal tubule cells alone is sufficient to inhibit UNX- or amino acid-induced hypertrophic nephron growth. Furthermore, our mechanistic studies unveiled that the SLC34a1-CreERT2 recombinase-mediated Pik3c3 downregulation inhibited UNX- or amino acid-stimulated lysosomal localization and signaling activation of mechanistic target of rapamycin complex 1 (mTORC1) in the renal proximal tubules. Moreover, our additional cell culture experiments using RNAi confirmed that knocking down Pik3c3 expression inhibited amino acid-stimulated mTORC1 signaling and blunted cellular growth in primary cultures of renal proximal tubule cells. Together, both our in vivo and in vitro experimental results indicate that Pik3c3 is a major mechanistic mediator responsible for sensing amino acid availability and initiating hypertrophic growth of renal proximal tubule cells by activation of the mTORC1-S6K1-rpS6 signaling pathway.


Assuntos
Classe III de Fosfatidilinositol 3-Quinases/genética , Túbulos Renais Proximais/crescimento & desenvolvimento , Rim/efeitos dos fármacos , Néfrons/crescimento & desenvolvimento , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Animais , Classe III de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Integrases/genética , Rim/crescimento & desenvolvimento , Rim/patologia , Rim/cirurgia , Túbulos Renais Proximais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Nefrectomia , Néfrons/metabolismo , Fosforilação/genética , Proteína-Lisina 6-Oxidase/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia
13.
Neoreviews ; 20(12): e725-e736, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31792159

RESUMO

Preterm birth is associated with adverse renal health outcomes including hypertension, chronic kidney disease, and an increased rate of progression to end-stage renal failure. This review explores the antenatal, perinatal, and postnatal factors that affect the functional nephron mass of an individual and contribute to long-term kidney outcome. Health-care professionals have opportunities to increase their awareness of the risks to kidney health in this population. Optimizing maternal health around the time of conception and during pregnancy, providing kidney-focused supportive care in the NICU during postnatal nephrogenesis, and avoiding accelerating nephron loss throughout life may all contribute to improved long-term outcomes. There is a need for ongoing research into the long-term kidney outcomes of preterm survivors in mid-to-late adulthood as well as a need for further research into interventions that may improve ex utero nephrogenesis.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Hiperóxia/metabolismo , Néfrons/crescimento & desenvolvimento , Insuficiência Renal Crônica/epidemiologia , Causalidade , Progressão da Doença , Feminino , Taxa de Filtração Glomerular , Humanos , Hipertensão/epidemiologia , Recém-Nascido de Baixo Peso , Recém-Nascido , Recém-Nascido Prematuro , Rim/embriologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Falência Renal Crônica/epidemiologia , Nefrocalcinose/epidemiologia , Néfrons/embriologia , Néfrons/metabolismo , Tamanho do Órgão , Gravidez , Nascimento Prematuro , Efeitos Tardios da Exposição Pré-Natal
14.
Sci Rep ; 9(1): 15915, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31685872

RESUMO

The nephron is composed of distinct segments that perform unique physiological functions. Little is known about how multipotent nephron progenitor cells differentiate into different nephron segments. It is well known that ß-catenin signaling regulates the maintenance and commitment of mesenchymal nephron progenitors during kidney development. However, it is not fully understood how it regulates nephron segmentation after nephron progenitors undergo mesenchymal-to-epithelial transition. To address this, we performed ß-catenin loss-of-function and gain-of-function studies in epithelial nephron progenitors in the mouse kidney. Consistent with a previous report, the formation of the renal corpuscle was defective in the absence of ß-catenin. Interestingly, we found that epithelial nephron progenitors lacking ß-catenin were able to form presumptive proximal tubules but that they failed to further develop into differentiated proximal tubules, suggesting that ß-catenin signaling plays a critical role in proximal tubule development. We also found that epithelial nephron progenitors lacking ß-catenin failed to form the distal tubules. Expression of a stable form of ß-catenin in epithelial nephron progenitors blocked the proper formation of all nephron segments, suggesting tight regulation of ß-catenin signaling during nephron segmentation. This work shows that ß-catenin regulates the formation of multiple nephron segments along the proximo-distal axis of the mammalian nephron.


Assuntos
Rim/fisiologia , Néfrons/metabolismo , beta Catenina/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Mutação com Ganho de Função , Rim/crescimento & desenvolvimento , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Microtúbulos/metabolismo , Néfrons/crescimento & desenvolvimento , Néfrons/patologia , Organogênese , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo , beta Catenina/genética
15.
Am J Physiol Renal Physiol ; 317(5): F1318-F1330, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31509011

RESUMO

The incidence of diabetes mellitus has significantly increased among women of childbearing age, and it has been shown that prenatal exposure to maternal diabetes increases the risk of associated congenital anomalies of the kidney. Congenital anomalies of the kidney are among the leading causes of chronic kidney disease in children. To better understand the effect of maternal diabetes on kidney development, we analyzed wild-type offspring (DM_Exp) of diabetic Ins2+/C96Y mice (Akita mice). DM_Exp mice at postnatal day 34 have a reduction of ~20% in the total nephron number compared with controls, using the gold standard physical dissector/fractionator method. At the molecular level, the expression of the nephron progenitor markers sine oculis homeobox homolog 2 and Cited1 was increased in DM_Exp kidneys at postnatal day 2. Conversely, the number of early developing nephrons was diminished in DM_Exp kidneys. This was associated with decreased expression of the intracellular domain of Notch1 and the canonical Wnt target lymphoid enhancer binding factor 1. Together, these data suggest that the diabetic intrauterine environment impairs the differentiation of nephron progenitors into nephrons, possibly by perturbing the Notch and Wnt/ß-catenin signaling pathways.


Assuntos
Diabetes Gestacional , Insulina/genética , Néfrons/crescimento & desenvolvimento , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Feminino , Predisposição Genética para Doença , Genótipo , Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Mutação , Gravidez , Fatores de Transcrição/metabolismo
16.
J Tissue Eng Regen Med ; 13(9): 1724-1731, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31267702

RESUMO

Stem cell-derived organoids are emerging as sophisticated models for studying development and disease and as potential sources for developing organ substitutes. Unfortunately, although organoids containing renal structures have been generated from mouse and human pluripotent stem cells, there are still critical unanswered questions that are difficult to attain via in vitro systems, including whether these nonvascularized organoids have a stable and physiologically relevant phenotype or whether a suitable transplantation site for long-term in vivo studies can be identified. Even orthotopic engraftment of organoid cultures in the adult does not provide an environment conducive to vascularization and functional differentiation. Previously, we showed that the lymph node offers an alternative transplantation site where mouse metanephroi can differentiate into mature renal structures with excretory, homeostatic, and endocrine functions. Here, we show that the lymph node lends itself well as a niche to also grow human primary kidney rudiments and can additionally be viewed as a platform to interrogate emerging renal organoid cultures. Our study has a wide-ranging impact for tissue engineering approaches to rebuild functional tissues in vivo including-but not limited to-the kidney.


Assuntos
Linfonodos/crescimento & desenvolvimento , Modelos Biológicos , Néfrons/citologia , Néfrons/crescimento & desenvolvimento , Organogênese , Células-Tronco/citologia , Animais , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus
17.
Semin Cell Dev Biol ; 91: 147-152, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31178004

RESUMO

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.


Assuntos
Células Epiteliais/citologia , Glomerulosclerose Segmentar e Focal/fisiopatologia , Glomérulos Renais/citologia , Néfrons/citologia , Podócitos/citologia , Animais , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Glomérulos Renais/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Nicho de Células-Tronco , Células-Tronco/citologia
18.
Sci Rep ; 9(1): 8016, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31142767

RESUMO

To combat organ shortage in transplantation medicine, a novel strategy has been proposed to generate human organs from exogenous pluripotent stem cells utilizing the developmental mechanisms of pig embryos/foetuses. Genetically modified pigs missing specific organs are key elements in this strategy. In this study, we demonstrate the feasibility of using a genome-editing approach to generate anephrogenic foetuses in a genetically engineered pig model. SALL1 knockout (KO) was successfully induced by injecting genome-editing molecules into the cytoplasm of pig zygotes, which generated the anephrogenic phenotype. Extinguished SALL1 expression and marked dysgenesis of nephron structures were observed in the rudimentary kidney tissue of SALL1-KO foetuses. Biallelic KO mutations of the target gene induced nephrogenic defects; however, biallelic mutations involving small in-frame deletions did not induce the anephrogenic phenotype. Through production of F1 progeny from mutant founder pigs, we identified mutations that could reliably induce the anephrogenic phenotype and hence established a line of fertile SALL1-mutant pigs. Our study lays important technical groundwork for the realization of human kidney regeneration through the use of an empty developmental niche in pig foetuses.


Assuntos
Animais Geneticamente Modificados , Edição de Genes/métodos , Néfrons/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Fatores de Transcrição/genética , Aloenxertos/provisão & distribuição , Animais , Sistemas CRISPR-Cas/genética , Estudos de Viabilidade , Feminino , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Humanos , Transplante de Rim , Masculino , Mutação , Células-Tronco Pluripotentes/fisiologia , Regeneração/fisiologia , Sus scrofa , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Zigoto/crescimento & desenvolvimento
19.
Elife ; 82019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30758286

RESUMO

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.


Assuntos
Galectina 3/genética , Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Organogênese/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Rim/metabolismo , Lectinas Tipo C/genética , Macrófagos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/genética , Camundongos , Néfrons/metabolismo , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Células-Tronco/metabolismo
20.
J Pathol ; 247(1): 86-98, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30246301

RESUMO

Wilms tumour is a paediatric malignancy with features of halted kidney development. Here, we demonstrate that the Iroquois homeobox genes IRX3 and IRX5 are essential for mammalian nephrogenesis and govern the differentiation of Wilms tumour. Knock-out Irx3- /Irx5- mice showed a strongly reduced embryonic nephron formation. In human foetal kidney and Wilms tumour, IRX5 expression was already activated in early proliferative blastema, whereas IRX3 protein levels peaked at tubular differentiation. Accordingly, an orthotopic xenograft mouse model of Wilms tumour showed that IRX3-/- cells formed bulky renal tumours dominated by immature mesenchyme and active canonical WNT/ß-catenin-signalling. In contrast, IRX5-/- cells displayed activation of Hippo and non-canonical WNT-signalling and generated small tumours with abundant tubulogenesis. Our findings suggest that promotion of IRX3 signalling or inhibition of IRX5 signalling could be a route towards differentiation therapy for Wilms tumour, in which WNT5A is a candidate molecule for enforced tubular maturation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Proteínas de Homeodomínio/metabolismo , Neoplasias Renais/metabolismo , Néfrons/metabolismo , Fatores de Transcrição/metabolismo , Tumor de Wilms/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos Knockout , Morfogênese , Néfrons/crescimento & desenvolvimento , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Tumor de Wilms/genética , Tumor de Wilms/patologia , Via de Sinalização Wnt , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...