Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Kidney Int ; 103(1): 77-86, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36055600

RESUMO

The kidney is an essential organ that ensures bodily fluid homeostasis and removes soluble waste products from the organism. Nephrons, the functional units of the kidney, comprise a blood filter, the glomerulus or glomus, and an epithelial tubule that processes the filtrate from the blood or coelom and selectively reabsorbs solutes, such as sugars, proteins, ions, and water, leaving waste products to be eliminated in the urine. Genes coding for transporters are segmentally expressed, enabling the nephron to sequentially process the filtrate. The Xenopus embryonic kidney, the pronephros, which consists of a single large nephron, has served as a valuable model to identify genes involved in nephron formation and patterning. Therefore, the developmental patterning program that generates these segments is of great interest. Prior work has defined the gene expression profiles of Xenopus nephron segments via in situ hybridization strategies, but a comprehensive understanding of the cellular makeup of the pronephric kidney remains incomplete. Here, we carried out single-cell mRNA sequencing of the functional Xenopus pronephric nephron and evaluated its cellular composition through comparative analyses with previous Xenopus studies and single-cell mRNA sequencing of the adult mouse kidney. This study reconstructs the cellular makeup of the pronephric kidney and identifies conserved cells, segments, and associated gene expression profiles. Thus, our data highlight significant conservation in podocytes, proximal and distal tubule cells, and divergence in cellular composition underlying the capacity of each nephron to remove wastes in the form of urine, while emphasizing the Xenopus pronephros as a model for physiology and disease.


Assuntos
Rim , Néfrons , Animais , Camundongos , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Glomérulos Renais/embriologia , Néfrons/embriologia , RNA Mensageiro/genética , Xenopus laevis/embriologia
2.
Dev Biol ; 470: 62-73, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33197428

RESUMO

Recent advances in stem cell biology have enabled the generation of kidney organoids in vitro, and further maturation of these organoids is observed after experimental transplantation. However, the current organoids remain immature and their precise maturation stages are difficult to determine because of limited information on developmental stage-dependent gene expressions in the kidney in vivo. To establish relevant molecular coordinates, we performed single-cell RNA sequencing (scRNA-seq) on developing kidneys at different stages in the mouse. By selecting genes that exhibited upregulation at birth compared with embryonic day 15.5 as well as cell lineage-specific expression, we generated gene lists correlated with developmental stages in individual cell lineages. Application of these lists to transplanted embryonic kidneys revealed that most cell types, other than the collecting ducts, exhibited similar maturation to kidneys at the neonatal stage in vivo, revealing non-synchronous maturation across the cell lineages. Thus, our scRNA-seq data can serve as useful molecular coordinates to assess the maturation of developing kidneys and eventually of kidney organoids.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rim/crescimento & desenvolvimento , Rim/metabolismo , Animais , Animais Recém-Nascidos , Linhagem da Célula , Regulação para Baixo , Rim/citologia , Rim/embriologia , Glomérulos Renais/citologia , Glomérulos Renais/embriologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo , Transplante de Rim , Túbulos Renais/citologia , Túbulos Renais/embriologia , Túbulos Renais/crescimento & desenvolvimento , Túbulos Renais/metabolismo , Camundongos , Podócitos/citologia , Podócitos/metabolismo , RNA-Seq , Análise de Célula Única , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
3.
J Am Soc Nephrol ; 30(9): 1641-1658, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31405951

RESUMO

BACKGROUND: GATA3 is a dual-zinc finger transcription factor that regulates gene expression in many developing tissues. In the kidney, GATA3 is essential for ureteric bud branching, and mice without it fail to develop kidneys. In humans, autosomal dominant GATA3 mutations can cause renal aplasia as part of the hypoparathyroidism, renal dysplasia, deafness (HDR) syndrome that includes mesangioproliferative GN. This suggests that GATA3 may have a previously unrecognized role in glomerular development or injury. METHODS: To determine GATA3's role in glomerular development or injury, we assessed GATA3 expression in developing and mature kidneys from Gata3 heterozygous (+/-) knockout mice, as well as injured human and rodent kidneys. RESULTS: We show that GATA3 is expressed by FOXD1 lineage stromal progenitor cells, and a subset of these cells mature into mesangial cells (MCs) that continue to express GATA3 in adult kidneys. In mice, we uncover that GATA3 is essential for normal glomerular development, and mice with haploinsufficiency of Gata3 have too few MC precursors and glomerular abnormalities. Expression of GATA3 is maintained in MCs of adult kidneys and is markedly increased in rodent models of mesangioproliferative GN and in IgA nephropathy, suggesting that GATA3 plays a critical role in the maintenance of glomerular homeostasis. CONCLUSIONS: These results provide new insights on the role GATA3 plays in MC development and response to injury. It also shows that GATA3 may be a novel and robust nuclear marker for identifying MCs in tissue sections.


Assuntos
Fator de Transcrição GATA3/metabolismo , Glomerulonefrite/metabolismo , Glomérulos Renais/metabolismo , Animais , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA3/genética , Haploinsuficiência , Humanos , Glomérulos Renais/anormalidades , Glomérulos Renais/embriologia , Glomérulos Renais/patologia , Masculino , Células Mesangiais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Ratos , Ratos Wistar
4.
Am J Perinatol ; 36(10): 1072-1078, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30537771

RESUMO

OBJECTIVE: To verify the prevalence of systemic arterial hypertension (SAH) and to identify possible early predictors of SAH at ages 2 and 4 years in very low birth weight (VLBW) infants. STUDY DESIGN: This is a prospective cohort study including inborn children with birth weight (BW) <1,500 g. Arterial blood pressure measurements were performed at 2 and 4 years. Model 1 compared children with and those without SAH at age 4. Model 2 compared children who had SAH at ages 2 and 4 with the others. SAH was diagnosed if the systolic or/and diastolic pressures were above the 95th percentile. RESULTS: A total of 198 patients were included during the 5-year study period, of whom 56% had SAH at age 4. In model 1, white matter injury (WMI) and catch-up growth at age 2 were predictors of SAH at age 4. In model 2, bronchopulmonary dysplasia, WMI, catch-up growth at age 2, and BW were predictors of SAH at 2 and 4 years. SAH at age 2 was an independent risk factor for SAH at age 4. After a multivariate analysis of model 2, BW and catch-up growth were associated with SAH. CONCLUSION: Prevalence of SAH was high in VLBW infants; it was associated with low BW and catch-up growth at age 2.


Assuntos
Hipertensão , Recém-Nascido Prematuro , Recém-Nascido de muito Baixo Peso , Pré-Escolar , Feminino , Humanos , Recém-Nascido , Glomérulos Renais/embriologia , Masculino , Análise Multivariada , Prevalência , Estudos Prospectivos , Fatores de Risco
5.
Curr Opin Nephrol Hypertens ; 27(3): 159-164, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29432216

RESUMO

PURPOSE OF REVIEW: Earlier works of the glomerulogenesis described morphological steps and protein expression during in-vivo and in-vitro kidney development. Recent technologies using cell-specific or conditional knock-out mice for several factors provide important knowledge about cross-talk signaling among resident cells as local events. Based on the recent advancement, this review revisits comprehensive morphological development of the glomerulus. RECENT FINDINGS: Interactions of presumptive podocyte vascular endothelial growth factor with vascular endothelial growth factor-2 on angioblasts initiate glomerular vascularization. In induced pluripotent stem cells or organoid-derived nephron formation, the lack of endothelium and mesangial cells under differentiated podocytes suggests the presence of another unknown mechanism for glomerular neovascularization. Mesangial cell migration is prerequisite for glomerular looping by interaction of endothelial platelet-derived grothe factor beta and mesangial platelet-derived growth factor receptor beta and requires the coreceptor neuropilin1. Development of the filtration barrier is promoted by cross-talk among resident cells and may need shear stress. The components of the glomerular basement membrane change during glomerulogenesis, and endothelium and podocytes produce laminin and type IV collagen α1 and α2, whereas type IV collagen α3, α4, α5 is derived only from podocytes. SUMMARY: Glomerulogenesis progresses by dynamic cellular migration/differentiation induced by cross-talk signaling in resident cells. Glomerular vasculogenesis and subsequent capillary development provide insight into glomerular regeneration and remodeling for medical application.


Assuntos
Endotélio/fisiologia , Glomérulos Renais/embriologia , Glomérulos Renais/fisiologia , Animais , Comunicação Celular , Diferenciação Celular , Movimento Celular , Colágeno Tipo IV/metabolismo , Endotélio/metabolismo , Humanos , Glomérulos Renais/irrigação sanguínea , Laminina/metabolismo , Neovascularização Fisiológica , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
J Vasc Res ; 55(5): 278-286, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212835

RESUMO

BACKGROUND/AIMS: Intussusceptive angiogenesis (IA) is a dynamic process which contributes to vascular expansion and remodeling. Intraluminal pillars have long been the distinctive structural indicator of IA. However, the mechanism of their formation has not been fully elucidated. METHODS: Using light and electron microscopy, we studied intussusceptive vascular growth in the developing porcine metanephric kidney. RESULTS: We observed intraluminal pillars formed by endothelial cells in the vasculature of developing glomeruli. Their diameter was < 2.5 µm, consistent with the diameter of nascent pillars. TEM revealed that the majority of these pillars consisted only of endothelium. However, a central core of extracellular matrix (ECM) covered by endothelium, reminiscent of a more mature intussusceptive pillar, was also found in the lumen of a glomerular capillary. Perivascular cells or pericytes were not involved in the pillar structure during these stages of formation. CONCLUSION: This study shows ECM presence in a mature intussusceptive pillar without any perivascular cell involvement in the structure. This leads to the hypothesis that ECM deposition precedes the participation of these cells in the formation of intraluminal pillars during IA in porcine metanephric glomerular capillaries.


Assuntos
Capilares/embriologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/embriologia , Neovascularização Fisiológica , Animais , Capilares/ultraestrutura , Células Endoteliais/ultraestrutura , Matriz Extracelular/ultraestrutura , Idade Gestacional , Glomérulos Renais/ultraestrutura , Microscopia Eletrônica de Transmissão , Organogênese , Sus scrofa
7.
PLoS Genet ; 11(7): e1005349, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26147622

RESUMO

African Americans have a disproportionate risk for developing nephropathy. This disparity has been attributed to coding variants (G1 and G2) in apolipoprotein L1 (APOL1); however, there is little functional evidence supporting the role of this protein in renal function. Here, we combined genetics and in vivo modeling to examine the role of apol1 in glomerular development and pronephric filtration and to test the pathogenic potential of APOL1 G1 and G2. Translational suppression or CRISPR/Cas9 genome editing of apol1 in zebrafish embryos results in podocyte loss and glomerular filtration defects. Complementation of apol1 morphants with wild-type human APOL1 mRNA rescues these defects. However, the APOL1 G1 risk allele does not ameliorate defects caused by apol1 suppression and the pathogenicity is conferred by the cis effect of both individual variants of the G1 risk haplotype (I384M/S342G). In vivo complementation studies of the G2 risk allele also indicate that the variant is deleterious to protein function. Moreover, APOL1 G2, but not G1, expression alone promotes developmental kidney defects, suggesting a possible dominant-negative effect of the altered protein. In sickle cell disease (SCD) patients, we reported previously a genetic interaction between APOL1 and MYH9. Testing this interaction in vivo by co-suppressing both transcripts yielded no additive effects. However, upon genetic or chemical induction of anemia, we observed a significantly exacerbated nephropathy phenotype. Furthermore, concordant with the genetic interaction observed in SCD patients, APOL1 G2 reduces myh9 expression in vivo, suggesting a possible interaction between the altered APOL1 and myh9. Our data indicate a critical role for APOL1 in renal function that is compromised by nephropathy-risk encoding variants. Moreover, our interaction studies indicate that the MYH9 locus is also relevant to the phenotype in a stressed microenvironment and suggest that consideration of the context-dependent functions of both proteins will be required to develop therapeutic paradigms.


Assuntos
Apolipoproteínas/genética , Glomerulonefrite Membranosa/genética , Glomérulos Renais/patologia , Lipoproteínas HDL/genética , Proteínas Motores Moleculares/genética , Cadeias Pesadas de Miosina/genética , Animais , Apolipoproteína L1 , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Variação Genética/genética , Taxa de Filtração Glomerular/genética , Humanos , Glomérulos Renais/embriologia , Glomérulos Renais/crescimento & desenvolvimento , Microscopia Eletrônica de Transmissão , Morfolinos/genética , Peixe-Zebra
8.
Am J Physiol Renal Physiol ; 312(4): F702-F715, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28148530

RESUMO

Unbiased transcriptome profiling and functional genomics approaches have identified ubiquitin-specific protease 40 (USP40) as a highly specific glomerular transcript. This gene product remains uncharacterized, and its biological function is completely unknown. Here, we showed that mouse and rat glomeruli exhibit specific expression of the USP40 protein, which migrated at 150 kDa and was exclusively localized in the podocyte cytoplasm of the adult kidney. Double-labeling immunofluorescence staining and confocal microscopy analysis of fetal and neonate kidney samples revealed that USP40 was also expressed in the vasculature, including in glomerular endothelial cells at the premature stage. USP40 in cultured glomerular endothelial cells and podocytes was specifically localized to the intermediate filament protein nestin. In glomerular endothelial cells, immunoprecipitation confirmed actual protein-protein binding of USP40 with nestin, and USP40-small-interfering RNA transfection revealed significant reduction of nestin. In a rat model of minimal-change nephrotic syndrome, USP40 expression was apparently reduced, which was also associated with the reduction of nestin. Zebrafish morphants lacking Usp40 exhibited disorganized glomeruli with the reduction of the cell junction in the endothelium and foot process effacement in the podocytes. Permeability studies in these zebrafish morphants demonstrated a disruption of the selective glomerular permeability filter. These data indicate that USP40/Usp40 is a novel protein that might play a crucial role in glomerulogenesis and the glomerular integrity after birth through the modulation of intermediate filament protein homeostasis.


Assuntos
Taxa de Filtração Glomerular , Glomérulos Renais/enzimologia , Ubiquitina Tiolesterase/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genótipo , Células HEK293 , Humanos , Glomérulos Renais/embriologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Nefrose Lipoide/enzimologia , Nefrose Lipoide/genética , Nefrose Lipoide/fisiopatologia , Nestina/metabolismo , Permeabilidade , Fenótipo , Podócitos/enzimologia , Interferência de RNA , Ratos , Transfecção , Ubiquitina Tiolesterase/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
9.
Semin Cell Dev Biol ; 36: 50-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25128732

RESUMO

The renal vascular bed has a stereotypic architecture that is essential for the kidney's role in excreting metabolic waste and regulating the volume and composition of body fluids. The kidney's excretory functions are dependent on the delivery of the majority of renal blood flow to the glomerular capillaries, which filter plasma removing from it metabolic waste, as well as vast quantities of solutes and fluids. The renal tubules reabsorb from the glomerular filtrate solutes and fluids required for homeostasis, while the post-glomerular capillary beds return these essential substances back into the systemic circulation. Thus, the kidney's regulatory functions are dependent on the close proximity or alignment of the post-glomerular capillary beds with the renal tubules. This review will focus on our current knowledge of the mechanisms controlling the embryonic development of the renal vasculature. An understanding of this process is critical for developing novel therapies to prevent vessel rarefaction and will be essential for engineering renal tissues suitable for restoring kidney function to the ever-increasing population of patients with end stage renal disease.


Assuntos
Glomérulos Renais/irrigação sanguínea , Túbulos Renais/irrigação sanguínea , Rim/irrigação sanguínea , Rim/embriologia , Humanos , Nefropatias/metabolismo , Glomérulos Renais/citologia , Glomérulos Renais/embriologia , Túbulos Renais/embriologia , Neovascularização Fisiológica
10.
Semin Cell Dev Biol ; 36: 39-49, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25153928

RESUMO

The glomerulus represents a highly structured filtration unit, composed of glomerular endothelial cells, mesangial cells, podocytes and parietal epithelial cells. During glomerulogenesis an intricate network of signaling pathways involving transcription factors, secreted factors and cell-cell communication is required to guarantee accurate evolvement of a functional, complex 3-dimensional glomerular architecture. Here, we want to provide an overview on the critical steps and relevant signaling cascades of glomerular development.


Assuntos
Glomérulos Renais/embriologia , Organogênese/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Comunicação Celular/fisiologia , Diferenciação Celular , Células Endoteliais/citologia , Humanos , Glomérulos Renais/citologia , Proteínas com Homeodomínio LIM/metabolismo , Fator de Transcrição MafB/metabolismo , Células Mesangiais/citologia , Podócitos/citologia , Receptores Notch/metabolismo , Fatores de Transcrição/metabolismo , Proteínas WT1/metabolismo
11.
BMC Dev Biol ; 16(1): 30, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27582005

RESUMO

BACKGROUND: Wnt11 is a member of the Wnt family of secreted signals controlling the early steps in ureteric bud (UB) branching. Due to the reported lethality of Wnt11 knockout embryos in utero, its role in later mammalian kidney organogenesis remains open. The presence of Wnt11 in the emerging tubular system suggests that it may have certain roles later in the development of the epithelial ductal system. RESULTS: The Wnt11 knockout allele was backcrossed with the C57Bl6 strain for several generations to address possible differences in penetrance of the kidney phenotypes. Strikingly, around one third of the null mice with this inbred background survived to the postnatal stages. Many of them also reached adulthood, but urine and plasma analyses pointed out to compromised kidney function. Consistent with these data the tubules of the C57Bl6 Wnt11 (-/-) mice appeared to be enlarged, and the optical projection tomography indicated changes in tubular convolution. Moreover, the C57Bl6 Wnt11 (-/-) mice developed secondary glomerular cysts not observed in the controls. The failure of Wnt11 signaling reduced the expression of several genes implicated in kidney development, such as Wnt9b, Six2, Foxd1 and Hox10. Also Dvl2, an important PCP pathway component, was downregulated by more than 90 % due to Wnt11 deficiency in both the E16.5 and NB kidneys. Since all these genes take part in the control of UB, nephron and stromal progenitor cell differentiation, their disrupted expression may contribute to the observed anomalies in the kidney tubular system caused by Wnt11 deficiency. CONCLUSIONS: The Wnt11 signal has roles at the later stages of kidney development, namely in coordinating the development of the tubular system. The C57Bl6 Wnt11 (-/-) mouse generated here provides a model for studying the mechanisms behind tubular anomalies and glomerular cyst formation.


Assuntos
Glomérulos Renais/anormalidades , Túbulos Renais/anormalidades , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Glomérulos Renais/embriologia , Túbulos Renais/embriologia , Camundongos , Camundongos Knockout , Transdução de Sinais
12.
Development ; 140(22): 4565-73, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24154527

RESUMO

Kidneys remove unwanted substances from the body and regulate the internal body environment. These functions are carried out by specialized cells (podocytes) that act as a filtration barrier between the internal milieu and the outside world, and by a series of tubules and ducts that process the filtrate and convey it to the outside. In the kidneys of amniote vertebrates, the filtration (podocyte) and tubular functions are tightly integrated into functional units called nephrons. The specification of the podocyte and tubular components of amniote nephrons is currently not well understood. The present study investigates podocyte and tubule differentiation in the avian mesonephric kidney, and presents several findings that refine our understanding of the initial events of nephron formation. First, well before the first morphological or molecular signs of nephron formation, mesonephric mesenchyme can be separated on the basis of morphology and the expression of the transcription factor Pod1 into dorsal and ventral components, which can independently differentiate in culture along tubule and podocyte pathways, respectively. Second, canonical Wnt signals, which are found in the nephric duct adjacent to the dorsal mesonephric mesenchyme and later in portions of the differentiating nephron, strongly inhibit podocyte but not tubule differentiation, suggesting that Wnt signaling plays an important role in the segmentation of the mesonephric mesenchyme into tubular and glomerular segments. The results are discussed in terms of their broader implications for models of nephron segmentation.


Assuntos
Padronização Corporal , Galinhas/metabolismo , Túbulos Renais/embriologia , Túbulos Renais/metabolismo , Podócitos/citologia , Via de Sinalização Wnt , Animais , Diferenciação Celular , Embrião de Galinha , Glomérulos Renais/citologia , Glomérulos Renais/embriologia , Túbulos Renais/citologia , Mesoderma/citologia , Mesoderma/embriologia , Mesonefro/embriologia , Modelos Biológicos , Néfrons/citologia , Néfrons/metabolismo , Podócitos/metabolismo , Fatores de Tempo
13.
J Am Soc Nephrol ; 26(3): 576-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25145929

RESUMO

The planar cell polarity (PCP) signaling pathway is crucial for tissue morphogenesis. Van Gogh-like protein 2 (Vangl2) is central in the PCP pathway; in mice, Vangl2 loss is embryonically lethal because of neural tube defects, and mutations in Vangl2 are associated with human neural tube defects. In the kidney, PCP signaling may be important for tubular morphogenesis and organization of glomerular epithelial cells (podocytes) along the glomerular basement membrane. Podocyte cell protrusions (foot processes) are critical for glomerular permselectivity; loss of foot process architecture results in proteinuria and FSGS. Previously, we showed a profound effect of PCP signaling on podocyte shape, actin rearrangement, cell motility, and nephrin endocytosis. To test our hypothesis that the PCP pathway is involved in glomerular development and function and circumvent lethality of the ubiquitous Vangl2 mutation in the Looptail mouse, we generated a mouse model with a podocyte-specific ablation of the Vangl2 gene. We report here that podocyte-specific deletion of Vangl2 leads to glomerular maturation defects in fetal kidneys. In adult mice, we detected significantly smaller glomeruli, but it did not affect glomerular permselectivity in aging animals. However, in the context of glomerular injury induced by injection of antiglomerular basement membrane antibody, deletion of Vangl2 resulted in exacerbation of injury and accelerated progression to chronic segmental and global glomerular sclerosis. Our results indicate that Vangl2 function in podocytes is important for glomerular development and protects against glomerular injury in adult animals.


Assuntos
Polaridade Celular , Glomérulos Renais/embriologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Feminino , Glomérulos Renais/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfogênese , Podócitos/metabolismo
14.
Kidney Int ; 87(6): 1125-40, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25651362

RESUMO

MicroRNAs, activated by the enzyme Dicer1, control post-transcriptional gene expression. Dicer1 has important roles in the epithelium during nephrogenesis, but its function in stromal cells during kidney development is unknown. To study this, we inactivated Dicer1 in renal stromal cells. This resulted in hypoplastic kidneys, abnormal differentiation of the nephron tubule and vasculature, and perinatal mortality. In mutant kidneys, genes involved in stromal cell migration and activation were suppressed as were those involved in epithelial and endothelial differentiation and maturation. Consistently, polarity of the proximal tubule was incorrect, distal tubule differentiation was diminished, and elongation of Henle's loop attenuated resulting in lack of inner medulla and papilla in stroma-specific Dicer1 mutants. Glomerular maturation and capillary loop formation were abnormal, whereas peritubular capillaries, with enhanced branching and increased diameter, formed later. In Dicer1-null renal stromal cells, expression of factors associated with migration, proliferation, and morphogenic functions including α-smooth muscle actin, integrin-α8, -ß1, and the WNT pathway transcriptional regulator LEF1 were reduced. Dicer1 mutation in stroma led to loss of expression of distinct microRNAs. Of these, miR-214, -199a-5p, and -199a-3p regulate stromal cell functions ex vivo, including WNT pathway activation, migration, and proliferation. Thus, Dicer1 activity in the renal stromal compartment regulates critical stromal cell functions that, in turn, regulate differentiation of the nephron and vasculature during nephrogenesis.


Assuntos
Diferenciação Celular/genética , RNA Helicases DEAD-box/fisiologia , Neovascularização Fisiológica/genética , Néfrons/embriologia , Ribonuclease III/fisiologia , Actinas/metabolismo , Animais , Capilares/embriologia , Movimento Celular/genética , Proliferação de Células/genética , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Feminino , Expressão Gênica , Cadeias alfa de Integrinas/metabolismo , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/citologia , Glomérulos Renais/embriologia , Túbulos Renais/irrigação sanguínea , Túbulos Renais/citologia , Túbulos Renais/embriologia , Túbulos Renais Distais/irrigação sanguínea , Túbulos Renais Distais/citologia , Túbulos Renais Distais/embriologia , Túbulos Renais Proximais/irrigação sanguínea , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/embriologia , Alça do Néfron/irrigação sanguínea , Alça do Néfron/citologia , Alça do Néfron/embriologia , Camundongos , MicroRNAs/genética , Néfrons/anormalidades , Néfrons/citologia , Organogênese/genética , Podócitos/fisiologia , Ribonuclease III/genética , Ribonuclease III/metabolismo , Células Estromais/fisiologia , Transcriptoma , Ureter/anormalidades , Via de Sinalização Wnt/genética
15.
Kidney Blood Press Res ; 40(4): 413-23, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26227088

RESUMO

BACKGROUND/AIMS: Cyclosporine A (CsA) is a commonly used immunosuppressive agent. In some patients treatment with CsA has to be continued during pregnancy. The aim of the study was to assess in an experimental model whether the exposure to CsA during fetal life influences the number and volume of glomeruli, kidney function and blood pressure in the offspring. METHODS: Eight pregnant female Sprague-Dawley rats were allocated to 2 treatment regimens: with CsA or solvent. Blood pressure was measured in the offspring at 7 and 11 weeks of age and albuminuria was determined at 11 weeks of age. In the kidney the number and mean volume of glomeruli was assessed using stereological methods. RESULTS: In the offspring of pregnant rats treated with CsA the number of glomeruli was significantly lower and the mean volume of glomeruli was higher when compared to the offspring of pregnant rats receiving solvent. Systolic and diastolic blood pressures as well as albuminuria were significantly higher in the offspring of mothers treated with CsA during gestation compared to the offspring from the control group. CONCLUSIONS: Exposure of rats to CsA during fetal life impairs kidney development, thus potentially predisposing to chronic kidney disease and hypertension in the adult life.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Ciclosporina/toxicidade , Imunossupressores/toxicidade , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/embriologia , Adulto , Albuminúria/metabolismo , Animais , Peso ao Nascer/efeitos dos fármacos , Feminino , Humanos , Testes de Função Renal , Glomérulos Renais/crescimento & desenvolvimento , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Resultado da Gravidez , Ratos Sprague-Dawley
16.
J Am Soc Nephrol ; 25(11): 2459-70, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24904088

RESUMO

Podocytes are terminally differentiated cells with an elaborate cytoskeleton and are critical components of the glomerular barrier. We identified a bHLH transcription factor, Tcf21, that is highly expressed in developing and mature podocytes. Because conventional Tcf21 knockout mice die in the perinatal period with major cardiopulmonary defects, we generated a conditional Tcf21 knockout mouse to explore the role of this transcription factor in podocytes in vivo. Tcf21 was deleted from podocytes and podocyte progenitors using podocin-cre (podTcf21) and wnt4-cre (wnt4creTcf21) driver strains, respectively. Loss of Tcf21 from capillary-loop stage podocytes (podTcf21) results in simplified glomeruli with a decreased number of endothelial and mesangial cells. By 5 weeks of age, 40% of podTcf21 mice develop massive proteinuria and lesions similar to FSGS. Notably, the remaining 60% of mice do not develop proteinuria even when aged to 8 months. By contrast, earlier deletion of Tcf21 from podocyte precursors (wnt4creTcf21) results in a profound developmental arrest of podocyte differentiation and renal failure in 100% of mice during the perinatal period. Taken together, our results demonstrate a critical role for Tcf21 in the differentiation and maintenance of podocytes. Identification of direct targets of this transcription factor may provide new therapeutic avenues for proteinuric renal disease, including FSGS.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diabetes Mellitus Experimental/fisiopatologia , Glomerulosclerose Segmentar e Focal/fisiopatologia , Podócitos/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Senescência Celular/fisiologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Glomérulos Renais/embriologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Óperon Lac , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Podócitos/patologia , Proteinúria/genética , Proteinúria/patologia , Proteinúria/fisiopatologia
17.
J Biol Chem ; 288(33): 24035-48, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23824190

RESUMO

The noncanonical Wnt/planar cell polarity (PCP) pathway controls a variety of cell behaviors such as polarized protrusive cell activity, directional cell movement, and oriented cell division and is crucial for the normal development of many tissues. Mutations in the PCP genes cause malformation in multiple organs. Recently, the PCP pathway was shown to control endocytosis of PCP and non-PCP proteins necessary for cell shape remodeling and formation of specific junctional protein complexes. During formation of the renal glomerulus, the glomerular capillary becomes enveloped by highly specialized epithelial cells, podocytes, that display unique architecture and are connected via specialized cell-cell junctions (slit diaphragms) that restrict passage of protein into the urine; podocyte differentiation requires active remodeling of cytoskeleton and junctional protein complexes. We report here that in cultured human podocytes, activation of the PCP pathway significantly stimulates endocytosis of the core slit diaphragm protein, nephrin, via a clathrin/ß-arrestin-dependent endocytic route. In contrast, depletion of the PCP protein Vangl2 leads to an increase of nephrin at the cell surface; loss of Vangl2 functions in Looptail mice results in disturbed glomerular maturation. We propose that the PCP pathway contributes to podocyte development by regulating nephrin turnover during junctional remodeling as the cells differentiate.


Assuntos
Polaridade Celular , Endocitose , Proteínas de Membrana/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Arrestinas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Polaridade Celular/efeitos dos fármacos , Clatrina/metabolismo , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , Glomérulos Renais/embriologia , Glomérulos Renais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Podócitos/efeitos dos fármacos , Ratos , Proteínas Wnt/farmacologia , beta-Arrestinas
18.
Am J Physiol Renal Physiol ; 307(10): F1105-10, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25186294

RESUMO

Indomethacin and ibuprofen are administered to close a patent ductus arteriosus (PDA) during active glomerulogenesis. Light and electron microscopic glomerular changes with no change in glomerular number were seen following indomethacin and ibuprofen treatment during glomerulogenesis at 14 days after birth in a neonatal rat model. This present study aimed to determine whether longstanding renal structural changes are present at 30 days and 6 mo (equivalent to human adulthood). Rat pups were administered indomethacin or ibuprofen antenatally on days 18-20 (0.5 mg·kg(-1)·dose(-1) indomethacin; 10 mg·kg(-1)·dose(-1) ibuprofen) or postnatally intraperitoneally from day 1 to 3 or day 1 to 5 (0.2 mg·kg(-1)·dose(-1) indomethacin; 10 mg·kg(-1)·dose(-1) ibuprofen). Control groups received no treatment or normal saline intraperitoneally. Pups were killed at 30 days of age and 6 mo of age. Tissue blocks from right kidneys were prepared for light and electron microscopic examination, while total glomerular number was determined in left kidneys using unbiased stereology. Eight pups were included in each group from 14 maternal rats. At 30 days and 6 mo, there were persistent electron microscopy abnormalities of the glomerular basement membrane in those receiving postnatal indomethacin and ibuprofen. There were no significant light microscopy findings at 30 days or 6 mo. At 6 mo, there were significantly fewer glomeruli in those receiving postnatal indomethacin but not ibuprofen (P = 0.003). In conclusion, indomethacin administered during glomerulogenesis appears to reduce the number of glomeruli in adulthood. Alternative options for closing a PDA should be considered including ibuprofen as well as emerging therapies such as paracetamol.


Assuntos
Inibidores de Ciclo-Oxigenase/efeitos adversos , Ibuprofeno/efeitos adversos , Indometacina/efeitos adversos , Glomérulos Renais/efeitos dos fármacos , Tocolíticos/efeitos adversos , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Feminino , Glomérulos Renais/embriologia , Glomérulos Renais/ultraestrutura , Gravidez , Ratos Sprague-Dawley
19.
Biochem Biophys Res Commun ; 444(4): 473-9, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24472557

RESUMO

The PKD1 gene is essential for a number of biological functions, and its loss-of-function causes autosomal dominant polycystic kidney disease (ADPKD). The gene is developmentally regulated and believed to play an essential role in renal development. Previous studies have shown that manipulating murine renal organ cultures with dominant-negative forms of the Pkd1 gene impaired ureteric bud (UB) branching. In the current study, we analyzed different stages of renal development in two distinct mouse models carrying either a null mutation or inactivation of the last two exons of Pkd1. Surprisingly, metanephric explants from Pkd1-deleted kidneys harvested at day E11.5 did not show defects of UB branching and elongation, estimated by cytokeratin staining on fixed tissues or by Hoxb7-GFP time-lapse imaging. However, renal explants from Pkd1-mutants isolated at day E14.5 showed impaired nephrogenesis. Notably, we observed cell migratory defects in the developing endothelial compartment. Previous studies had implicated the Pkd1 gene in controlling cell migration and collagen deposition through PI3 kinases. In line with these studies, our results show that wild-type explants treated with PI3-kinase inhibitors recapitulate the endothelial defects observed in Pkd1 mutants, whereas treatment with VEGF only partially rescued the defects. Our data are consistent with a role for the Pkd1 gene in the endothelium that may be required for proper nephrogenesis.


Assuntos
Glomérulos Renais/embriologia , Glomérulos Renais/fisiopatologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Canais de Cátion TRPP/genética , Animais , Movimento Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Glomérulos Renais/metabolismo , Camundongos , Mutação , Técnicas de Cultura de Órgãos , Inibidores de Fosfoinositídeo-3 Quinase , Canais de Cátion TRPP/metabolismo
20.
Development ; 138(3): 465-74, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21205791

RESUMO

Despite extensive study of the development of the nephron, which is the functional unit of the kidney, the molecular mechanisms underlying the determination of nephron size remain largely unknown. Using the Xenopus pronephros, we demonstrate here that Tbx2, a T-box transcriptional repressor, functions to demarcate the territory of the pronephric nephron. Tbx2 is specifically expressed around three distinct components of the pronephric nephron: the tubule, duct and glomus. Gain of function of Tbx2 inhibits nephric mesoderm formation. Conversely, Tbx2 loss of function expands the boundary of each component of the pronephric nephron, resulting in an enlarged pronephros. BMP signals induce Tbx2 in the non-nephric mesoderm, which inhibits the expression of the nephric markers Hey1 and Gremlin. Importantly, these pronephric molecules repress Tbx2 expression by antagonizing BMP signals in the nephric mesoderm. These results suggest that the negative regulatory loops between BMP/Tbx2 and Gremlin or Hey1 are responsible for defining the territory of the pronephric nephron.


Assuntos
Néfrons/metabolismo , Proteínas com Domínio T/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Citocinas , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glomérulos Renais/embriologia , Glomérulos Renais/metabolismo , Túbulos Renais/embriologia , Túbulos Renais/metabolismo , Néfrons/embriologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas com Domínio T/genética , Proteínas de Xenopus/genética , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA