Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
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.
Nat Commun ; 10(1): 5705, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31836710

RESUMO

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Assuntos
Capilares/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glomérulos Renais/irrigação sanguínea , Animais , Capilares/citologia , Capilares/metabolismo , Células Cultivadas , Embrião de Mamíferos , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Perfilação da Expressão Gênica , Humanos , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Cultura Primária de Células , RNA-Seq , Análise de Célula Única , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
4.
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
5.
Pediatr Res ; 85(5): 724-730, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30700837

RESUMO

BACKGROUND: Renin-angiotensin system (RAS) blockade during nephrogenesis causes a broad range of renal mal-development. Here, we hypothesized that disruption of renal lymphangiogenesis may contribute to tubulointerstitial alterations after RAS blockade during kidney maturation. METHODS: Newborn rat pups were treated with enalapril (30 mg/kg/day) or vehicle for 7 days after birth. Lymphangiogenesis was assessed via immunostaining and/or immunoblots for vascular endothelial growth factor (VEGF)-C, VEGF receptor (VEGFR)-3, Podoplanin, and Ki-67. The intrarenal expression of fibroblast growth factor (FGF)-1, FGF-2, FGF receptor (R)-1, α-smooth muscle actin (α-SMA), and fibroblast-specific protein (FSP)-1 was also determined. Sirius Red staining was performed to evaluate interstitial collagen deposition. RESULTS: On postnatal day 8, renal lymphangiogenesis was disrupted by neonatal enalapril treatment. The expression of podoplanin and Ki-67 decreased in enalapril-treated kidneys. While the expression of VEGF-C was decreased, the levels of VEGFR-3 receptor increased following enalapril treatment. Enalapril treatment also reduced the renal expression of FGF-1, FGF-2, and FGFR-1. Enalapril-treated kidneys exhibited profibrogenic properties with increased expression of α-SMA and FSP-1 and enhanced deposition of interstitial collagen. CONCLUSION: Enalapril treatment during postnatal renal maturation can disrupt renal lymphangiogenesis along with tubulointerstitial changes, which may result in a pro-fibrotic environment in the developing rat kidney.


Assuntos
Angiotensinas/antagonistas & inibidores , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/metabolismo , Colágeno/metabolismo , Enalapril/farmacologia , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibrose , Antígeno Ki-67/metabolismo , Nefropatias/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Túbulos Renais/crescimento & desenvolvimento , Túbulos Renais/patologia , Linfangiogênese , Músculo Liso/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Matrix Biol ; 75-76: 58-71, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29803937

RESUMO

Basement membranes (BMs) are thin dense sheets of extracellular matrix that surround most tissues. When the BMs of neighboring tissues come into contact, they usually slide along one another and act to separate tissues and organs into distinct compartments. However, in certain specialized regions, the BMs of neighboring tissues link, helping to bring tissues together. These BM connections can be transient, such as during tissue fusion events in development, or long-term, as with adult tissues involved with filtration, including the blood brain barrier and kidney glomerulus. The transitory nature of these connections in development and the complexity of tissue filtration systems in adults have hindered the understanding of how juxtaposed BMs fasten together. The recent identification of a BM-BM adhesion system in C. elegans, termed B-LINK (BM linkage), however, is revealing cellular and extracellular matrix components of a nascent tissue adhesion system. We discuss insights gained from studying the B-LINK tissue adhesion system in C. elegans, compare this adhesion with other BM-BM connections in Drosophila and vertebrates, and outline important future directions towards elucidating this fascinating and poorly understood mode of adhesion that joins neighboring tissues.


Assuntos
Membrana Basal/metabolismo , Matriz Extracelular/genética , Aderências Teciduais/genética , Animais , Membrana Basal/crescimento & desenvolvimento , Barreira Hematoencefálica/crescimento & desenvolvimento , Barreira Hematoencefálica/metabolismo , Caenorhabditis elegans/genética , Comunicação Celular/genética , Compartimento Celular/genética , Drosophila/genética , Matriz Extracelular/metabolismo , Humanos , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo
7.
Sci Rep ; 8(1): 14723, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30283057

RESUMO

Kidney-derived c-kit+ cells exhibit progenitor/stem cell properties and can regenerate epithelial tubular cells following ischemia-reperfusion injury in rats. We therefore investigated whether c-kit+ progenitor/stem cells contribute to podocyte repair in a rat model of acute proteinuria induced by puromycin aminonucleoside (PAN), the experimental prototype of human minimal change disease and early stages of focal and segmental glomerulosclerosis. We found that c-kit+ progenitor/stem cells accelerated kidney recovery by improving foot process effacement (foot process width was lower in c-kit group vs saline treated animals, P = 0.03). In particular, these cells engrafted in small quantity into tubules, vessels, and glomeruli, where they occasionally differentiated into podocyte-like cells. This effect was related to an up regulation of α-Actinin-4 and mTORC2-Rictor pathway. Activation of autophagy by c-kit+ progenitor/stem cells also contributed to kidney regeneration and intracellular homeostasis (autophagosomes and autophagolysosomes number and LC3A/B-I and LC3A/B-II expression were higher in the c-kit group vs saline treated animals, P = 0.0031 and P = 0.0009, respectively). Taken together, our findings suggest that kidney-derived c-kit+ progenitor/stem cells exert reparative effects on glomerular disease processes through paracrine effects, to a lesser extent differentiation into podocyte-like cells and contribution to maintenance of podocyte cytoskeleton after injury. These findings have clinical implications for cell therapy of glomerular pathobiology.


Assuntos
Podócitos/metabolismo , Proteinúria/genética , Proteínas Proto-Oncogênicas c-kit/genética , Regeneração/genética , Actinina/genética , Animais , Diferenciação Celular/genética , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Rim/metabolismo , Rim/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Nefrose Lipoide , Proteinúria/induzido quimicamente , Proteinúria/patologia , Puromicina Aminonucleosídeo/toxicidade , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Células-Tronco/metabolismo
8.
Nephron ; 138(4): 310-323, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29342457

RESUMO

BACKGROUND: von Hippel-Lindau (VHL) disease is characterized by the development of benign and malignant tumours in many organ systems, including renal cysts and clear cell renal cell carcinoma. It is not completely understood what underlies the development of renal pathology, and the use of murine Vhl models has been challenging due to limitations in disease conservation. We previously described a zebrafish model bearing inactivating mutations in the orthologue of the human VHL gene. METHODS: We used histopathological and functional assays to investigate the pronephric and glomerular developmental defects in vhl mutant zebrafish, supported by human cell culture assays. RESULTS: Here, we report that vhl is required to maintain pronephric tubule and glomerulus integrity in zebrafish embryos. vhl mutant glomeruli are enlarged, cxcr4a+ capillary loops are dilated and the Bowman space is widened. While we did not observe pronephric cysts, the cells of the proximal convoluted and anterior proximal straight tubule are enlarged, periodic acid schiff (PAS) and Oil Red O positive, and display a clear cytoplasm after hematoxylin and eosine staining. Ultrastructural analysis showed the vhl-/- tubule to accumulate large numbers of vesicles of variable size and electron density. Microinjection of the endocytic fluorescent marker AM1-43 in zebrafish embryos revealed an accumulation of endocytic vesicles in the vhl mutant pronephric tubule, which we can recapitulate in human cells lacking VHL. CONCLUSIONS: Our data indicates that vhl is required to maintain pronephric tubule and glomerulus integrity during zebrafish development, and suggests a role for VHL in endocytic vesicle trafficking.


Assuntos
Glomérulos Renais/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Animais , Desenvolvimento Embrionário/genética , Glomérulos Renais/anormalidades , Glomérulos Renais/crescimento & desenvolvimento , Túbulos Renais Proximais/anormalidades , Túbulos Renais Proximais/crescimento & desenvolvimento , Larva , Mutação , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
9.
JCI Insight ; 2(2): e88848, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28138555

RESUMO

The neonatal mouse kidney retains nephron progenitor cells in a nephrogenic zone for 3 days after birth. We evaluated whether de novo nephrogenesis can be induced postnatally beyond 3 days. Given the long-term implications of nephron number for kidney health, it would be useful to enhance nephrogenesis in the neonate. We induced nephron reduction by cryoinjury with or without contralateral nephrectomy during the neonatal period or after 1 week of age. There was no detectable compensatory de novo nephrogenesis, as determined by glomerular counting and lineage tracing. Contralateral nephrectomy resulted in additional adaptive healing, with little or no fibrosis, but did not also stimulate de novo nephrogenesis. In contrast, injury initiated at 1 week of age led to healing with fibrosis. Thus, despite the presence of progenitor cells and ongoing nephron maturation in the newborn mouse kidney, de novo nephrogenesis is not inducible by acute nephron reduction. This indicates that additional nephron progenitors cannot be recruited after birth despite partial renal ablation providing a reparative stimulus and suggests that nephron number in the mouse is predetermined at birth.


Assuntos
Criocirurgia , Nefrectomia , Néfrons/crescimento & desenvolvimento , Células-Tronco , Animais , Animais Recém-Nascidos , Linhagem da Célula , Fibrose , Proteínas de Homeodomínio/metabolismo , Rim/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Néfrons/patologia , Organogênese , Fator de Transcrição PAX2/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma
10.
Exp Anim ; 66(3): 183-189, 2017 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-28179596

RESUMO

Newborn mouse glomeruli are still immature with a morphological feature of an early capillary loop stage, but infant mice do not manifest proteinuria. Little is known about the molecular mechanism whereby infant mice are resistant to proteinuria. Nephrin and synaptopodin are crucial for slit diaphragm and foot process (FP) formation for avoiding proteinuria. Nephrin tyrosine phosphorylation means a transient biological signaling required for FP repair or extension during nephrotic disease. Using an immunohistochemical technique, we examined the natural course of nephrin, Wilms' tumor-1 (WT1) and synaptopodin at 16.5 days of embryonic age (E16.5d) and E19.5d, 7 days of post-neonatal age (P7d) and P42d during renal development of mice. As a result, nephrin and synaptopodin were detected at E19.5d in S-shaped bodies. WT1, a transcriptional factor for nephrin, was detected in nucleus in podocyte-like cells in all stages. Nephrin tyrosine phosphorylation was evident in glomeruli at P7d, and this was associated with an early-stage of FP extension. Inversely, nephrin phosphorylation became faint at P42d, along with maturated FP. Based on the present results, we suggest the sequential molecular mechanism to protect growing mice from proteinuria: (i) WT1-induced nephrin production by podocytes in S-shaped bodies at E19.5d; (ii) Synchronized induction of synaptopodin at the same period; and (iii) FP extension is initiated at a milk-suckling stage under a nephrin tyrosine-phosphorylated condition, while it is arrested at an adult stage, associated with a loss of nephrin-based signaling.


Assuntos
Expressão Gênica , Glomérulos Renais/embriologia , Glomérulos Renais/crescimento & desenvolvimento , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Podócitos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Animais , Animais Recém-Nascidos , Glomérulos Renais/citologia , Camundongos Endogâmicos C57BL , Proteínas WT1
11.
Physiol Rep ; 4(16)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27796270

RESUMO

The influence of sleep restriction (SR) during pregnancy on blood pressure and renal function among female adult offspring was investigated. Pregnant Wistar rats were distributed into control and SR groups. The SR was performed between the 14th and 20th days of pregnancy (multiple platforms method for 20 h/day). At 2 months of age, half of the offspring from both groups were subjected to an ovariectomy (ovx), and the other half underwent sham surgery. The groups were as follows: control sham (Csham), control ovx (Covx), SR sham (SRsham), and SR ovx (SRovx). Renal function markers and systolic blood pressure (BPi, indirect method) were evaluated at 4, 6, and 8 months. Subsequently, the rats were euthanized, kidneys were removed, and processed for morphological analyses of glomerular area (GA), number of glomeruli per mm3 (NG), and kidney mass (KM). Increased BPi was observed in the Covx, SRsham, and SRovx groups compared to Csham at all ages. Increased plasma creatinine concentration and decreased creatinine clearance were observed in the SRsham and SRovx groups compared to the Csham and Covx groups. The SRovx group showed higher BPi and reduced creatinine clearance compared to all other groups. The SRovx group showed reduced values of GA and KM, as well as increased NG, macrophage infiltration, collagen deposit, and ACE1 expression at the renal cortex. Therefore, SR during pregnancy might be an additional risk factor for developing renal dysfunction and increasing BP in female adult offspring. The absence of female hormones exacerbates the changes caused by SR.


Assuntos
Pressão Sanguínea/fisiologia , Taxa de Filtração Glomerular/fisiologia , Hipertensão/etiologia , Nefropatias/etiologia , Rim/citologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Privação do Sono/complicações , Animais , Animais Recém-Nascidos , Determinação da Pressão Arterial/métodos , Creatinina/sangue , Feminino , Desenvolvimento Fetal/fisiologia , Hipertensão/fisiopatologia , Rim/metabolismo , Nefropatias/fisiopatologia , Glomérulos Renais/crescimento & desenvolvimento , Tamanho do Órgão , Ovariectomia/efeitos adversos , Ovariectomia/métodos , Placebos/efeitos adversos , Gravidez , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Ratos Wistar , Fatores de Risco , Privação do Sono/metabolismo , Privação do Sono/fisiopatologia
12.
Nephron ; 132(2): 110-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26807737

RESUMO

BACKGROUND: Diuretics are administered to neonates to control fluid balance. We studied whether clinical doses affected kidney development and function and whether extrauterine growth retardation (EUGR) could be a modulator. METHODS: Wistar rats were cross-fostered in normal food or food restricted litters at postnatal day (PND) 2 and treated daily with 0.9% NaCl, 5 mg/kg furosemide or 5 mg/kg hydrochlorothiazide (HCTZ) up to PND 8. Kidneys were evaluated on proliferation, apoptosis and a set of mRNA target genes at PND 8, glomerular- and glomerular generation count at PND 35, clinical pathology parameters at 3- and 9 months, neutrophil gelatinase-associated lipocalin at PND 8, 3 and 6 months, monthly blood pressure from 3 months onward and histopathology at study end. RESULTS: Treatment with furosemide or HCTZ did not have relevant effects on measured parameters. EUGR resulted in lower body weight from day 3 onwards (-29% at weaning; p < 0.001, -10% at necropsy; p < 0.001), less glomerular generations (4.4 ± 0.32 vs. 5.0 ± 0.423; p = 0.025, males only), decreased glomerular numbers (27,861 ± 3,468 vs. 30,527 ± 4,096; p = 0.026), higher creatinine clearance (0.84 ± 0.1 vs. 0.77 ± 0.09 ml/min/kg; p = 0.047) at 3 months and lower plasma creatinine (25.7 ± 1.8 vs. 27.5 ± 2.8 µmol/l; p = 0.043) at 9 months. CONCLUSION: Furosemide and HCTZ did not influence kidney development or function when administered in a clinically relevant dose to rat pups at a stage of ongoing nephrogenesis. EUGR led to impaired kidney development but did not modify furosemide or HCTZ findings.


Assuntos
Diuréticos/farmacologia , Rim/efeitos dos fármacos , Rim/crescimento & desenvolvimento , Proteínas de Fase Aguda/metabolismo , Envelhecimento/metabolismo , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Furosemida/farmacologia , Hidroclorotiazida/farmacologia , Rim/fisiologia , Glomérulos Renais/citologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/crescimento & desenvolvimento , Lipocalina-2 , Lipocalinas/metabolismo , Masculino , Gravidez , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Cloreto de Sódio/farmacologia
13.
Am J Physiol Renal Physiol ; 307(2): F149-58, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24899060

RESUMO

Worldwide, approximately 10% of neonates are born preterm. The majority of preterm neonates are born when the kidneys are still developing; therefore, during the early postnatal period renal function is likely reflective of renal immaturity and/or injury. This study evaluated glomerular and tubular function and urinary neutrophil gelatinase-associated lipocalin (NGAL; a marker of renal injury) in preterm neonates during the first month of life. Preterm and term infants were recruited from Monash Newborn (neonatal intensive care unit at Monash Medical Centre) and Jesse McPherson Private Hospital, respectively. Infants were grouped according to gestational age at birth: ≤28 wk (n = 33), 29-31 wk (n = 44), 32-36 wk (n = 32), and term (≥37 wk (n = 22)). Measures of glomerular and tubular function were assessed on postnatal days 3-7, 14, 21, and 28. Glomerular and tubular function was significantly affected by gestational age at birth, as well as by postnatal age. By postnatal day 28, creatinine clearance remained significantly lower among preterm neonates compared with term infants; however, sodium excretion was not significantly different. Pathological proteinuria and high urinary NGAL levels were observed in a number of neonates, which may be indicative of renal injury; however, there was no correlation between the two markers. Findings suggest that neonatal renal function is predominantly influenced by renal maturity, and there was high capacity for postnatal tubular maturation among preterm neonates. There is insufficient evidence to suggest that urinary NGAL is a useful marker of renal injury in the preterm neonate.


Assuntos
Injúria Renal Aguda/fisiopatologia , Recém-Nascido Prematuro , Glomérulos Renais/fisiopatologia , Túbulos Renais/fisiopatologia , Injúria Renal Aguda/sangue , Injúria Renal Aguda/urina , Proteínas de Fase Aguda/urina , Fatores Etários , Biomarcadores/sangue , Biomarcadores/urina , Creatinina/sangue , Creatinina/urina , Idade Gestacional , Taxa de Filtração Glomerular , Humanos , Lactente Extremamente Prematuro , Recém-Nascido , Glomérulos Renais/crescimento & desenvolvimento , Túbulos Renais/crescimento & desenvolvimento , Lipocalina-2 , Lipocalinas/urina , Modelos Biológicos , Proteinúria/fisiopatologia , Proteinúria/urina , Proteínas Proto-Oncogênicas/urina , Vitória
14.
J Am Soc Nephrol ; 25(9): 1966-78, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24652804

RESUMO

TGF-ß-activated kinase 1 (TAK1) is a key intermediate in signal transduction induced by TGF-ß or inflammatory cytokines, such as TNF-α and IL-1, which are potent inducers of podocyte injury responses that lead to proteinuria and glomerulosclerosis. Nevertheless, little is known about the physiologic and pathologic roles of TAK1 in podocytes. To examine the in vivo role of TAK1, we generated podocyte-specific Tak1 knockout mice (Nphs2-Cre(+):Tak1(fx/fx); Tak1(∆/∆)). Targeted deletion of Tak1 in podocytes resulted in perinatal lethality, with approximately 50% of animals dying soon after birth and 90% of animals dying within 1 week of birth. Tak1(∆/∆) mice developed proteinuria from P1 and exhibited delayed glomerulogenesis and reduced expression of Wilms' tumor suppressor 1 and nephrin in podocytes. Compared with Tak1(fx/fx) mice, Tak1(∆/∆) mice exhibited impaired formation of podocyte foot processes that caused disruption of the podocyte architecture with prominent foot process effacement. Intriguingly, Tak1(∆/∆) mice displayed increased expression of vascular endothelial growth factor within the glomerulus and abnormally enlarged glomerular capillaries. Furthermore, 4- and 7-week-old Tak1(∆/∆) mice with proteinuria had increased collagen deposition in the mesangium and the adjacent tubulointerstitial area. Thus, loss of Tak1 in podocytes is associated with the development of proteinuria and glomerulosclerosis. Taken together, our data show that TAK1 regulates the expression of Wilms' tumor suppressor 1, nephrin, and vascular endothelial growth factor and that TAK1 signaling has a crucial role in podocyte differentiation and attainment of normal glomerular microvasculature during kidney development and glomerular filtration barrier homeostasis.


Assuntos
Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Podócitos/citologia , Podócitos/enzimologia , Animais , Animais Recém-Nascidos , Capilares/enzimologia , Capilares/crescimento & desenvolvimento , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Colágeno/metabolismo , Feminino , Barreira de Filtração Glomerular/irrigação sanguínea , Barreira de Filtração Glomerular/enzimologia , Barreira de Filtração Glomerular/crescimento & desenvolvimento , Glomérulos Renais/crescimento & desenvolvimento , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteinúria/enzimologia , Proteinúria/etiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo
15.
J Vet Med Sci ; 76(2): 277-80, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24107464

RESUMO

In this study, age-dependent histological changes in the kidneys of Brown Norway rat, a strain useful for conducting aging research, were evaluated. Examination was performed at 3, 12, 18, 24 and 30 months of age. Sclerotic and hypertrophic changes of the glomeruli were observed, and quantitative scores of these changes persistently increased with age. A marginal increase in scores was observed for glomerular cystic changes and tubulointerstitial damage. Further, urothelial hyperplasia was observed in the renal papillae, particularly at 30 months of age. In conclusion, the findings of the present study demonstrate that the Brown Norway strain exhibits persistent, but mild progression of age-dependent renal histological changes.


Assuntos
Envelhecimento/fisiologia , Glomérulos Renais/patologia , Rim/anatomia & histologia , Animais , Rim/fisiologia , Glomérulos Renais/crescimento & desenvolvimento , Ratos
16.
Kidney Int ; 85(5): 1091-102, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24172684

RESUMO

The coordination of multiple cytokines and transcription factors with their downstream signaling pathways has been shown to be integral to nephron maturation. Here we present a completely novel role for the helix-loop-helix transcription factor Early B-cell factor 1 (Ebf1), originally identified for B-cell maturation, for the proper maturation of glomerular cells from mesenchymal progenitors. The expression of Ebf1 was both spatially and temporally regulated within the developing cortex and glomeruli. Using Ebf1-null mice, we then identified biochemical, metabolic, and histological abnormalities in renal development that arose in the absence of this transcription factor. In the Ebf1 knockout mice, the developed kidneys show thinned cortices and reduced glomerular maturation. The glomeruli showed abnormal vascularization and severely effaced podocytes. The mice exhibited early albuminuria and elevated blood urea nitrogen levels. Moreover, the glomerular filtration rate was reduced >66% and the expression of podocyte-derived vascular endothelial growth factor A was decreased compared with wild-type control mice. Thus, Ebf1 has a significant and novel role in glomerular development, podocyte maturation, and the maintenance of kidney integrity and function.


Assuntos
Glomérulos Renais/metabolismo , Transativadores/metabolismo , Fatores Etários , Albuminúria/genética , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Animais , Nitrogênio da Ureia Sanguínea , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Taxa de Filtração Glomerular , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/fisiopatologia , Glomérulos Renais/ultraestrutura , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese , Fenótipo , Podócitos/metabolismo , Podócitos/ultraestrutura , Transdução de Sinais , Fatores de Tempo , Transativadores/deficiência , Transativadores/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Nephrol ; 24(4): 474-81, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20954140

RESUMO

INTRODUCTION: We analyzed renal histologic and immunohistologic findings in children with nephrotic syndrome (NS) who did (n=5) or did not (n=17) develop cyclosporine A (CyA) nephropathy despite appropriately low serum CyA concentrations being maintained over 2 years. METHODS: To discriminate embryonic-type from mature glomeruli, we performed staining for type IV collagen a1, laminin ß1 and laminin ß2. Staining patterns were used to semiquantitatively assess glomerular immaturity (glomerular immaturity index, or GII). RESULTS: In follow-up biopsy specimens, residual embryonic-type, collapsed embryonic-type and sclerotic glomeruli that had failed to differentiate were observed. Patients with early-onset CyA nephropathy had a high GII. In patients with a high GII, arteriopathy developed early in CyA treatment. Arteriopathy was observed mostly near embryonic-type glomeruli. Taken together, these glomeruli (surviving embryonic-type, collapsing embryonic-type, and sclerotic glomeruli) essentially equaled the total number of embryonic-type glomeruli in specimens obtained before CyA treatment. CONCLUSION: Our findings indicate a need for caution in CyA therapy for patients with NS, even for a relatively short course of administration, because some patients may have embryonic-type glomeruli or immature arterioles that predispose them to CyA nephropathy.


Assuntos
Ciclosporina/farmacologia , Imunossupressores/farmacologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/crescimento & desenvolvimento , Síndrome Nefrótica/patologia , Pré-Escolar , Colágeno Tipo IV/metabolismo , Ciclosporina/uso terapêutico , Humanos , Imuno-Histoquímica , Imunossupressores/uso terapêutico , Lactente , Glomérulos Renais/patologia , Laminina/metabolismo , Masculino , Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/metabolismo , Índice de Gravidade de Doença , Estatísticas não Paramétricas
18.
Cell Tissue Res ; 341(1): 197-209, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20495826

RESUMO

Most tubular epithelial cell types express primary cilia, and mutations of primary-cilium-associated proteins are well known to cause several kinds of cystic renal disease. However, until now, it has been unclear whether mammalian podocytes express primary cilia in vivo. In this study, we determined whether primary cilia are present in the podocytes of rat immature and mature glomeruli by means of transmission electron microscopy of serial ultrathin sections. In immature glomeruli of fetal rats, podocytes express the primary cilia with high percentages at the S-shaped body (88 +/- 5%, n = 3), capillary loop (95 +/- 4%, n = 4), and maturing glomerulus (76 +/- 13%, n = 5) stages. The percentage of ciliated podocytes was significantly lower at the maturing glomerulus stage than at the former two stages. In mature glomeruli of adult rats, ciliated podocytes were not found at all (0 +/- 0%, n = 11). These findings indicate that the primary cilia gradually disappear in rat podocytes during glomerular development. Since glomerular filtration rate increases during development, the primary cilia on the podocytes are subjected to a stronger bending force. Thus, the disappearance of the primary cilia presumably prevents the entry of excessive calcium-ions via the cilium-associated polycystin complexes and the disturbance of intracellular signaling cascades in mature podocytes.


Assuntos
Cílios/metabolismo , Glomérulos Renais/crescimento & desenvolvimento , Podócitos/metabolismo , Animais , Centríolos/ultraestrutura , Cílios/ultraestrutura , Feminino , Imunofluorescência , Imuno-Histoquímica , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Glomérulos Renais/ultraestrutura , Masculino , Podócitos/citologia , Podócitos/ultraestrutura , Ratos , Ratos Wistar
19.
Pediatr Int ; 52(3): 386-92, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19761519

RESUMO

BACKGROUND: A mouse model of impaired renal development was developed and the effect of retinoic acid (RA) was investigated in this animal model. METHODS: An angiogenesis inhibitor (SU1498) was injected s.c. into day 3 C57BL/6 newborn mice to create a model of arrested renal development. RA (2 mg/kg) was injected i.p. for 10 days. Morphometry and immunohistochemistry were done. RESULTS: Mice injected with SU1498 demonstrated deranged renal development in tubular structure and glomerular tuft area. Cortical thickness and area of glomerular tuft were significantly decreased after vascular endothelial growth factor (VEGF) inhibitor, and were significantly restored by RA. The length of capillary loops/glomerulus, the number of podocytes/glomerulus, and density of peritubular capillaries on CD31 immunostaining were significantly decreased by VEGF blocking and recovered by RA. CONCLUSIONS: VEGF plays a major role in renal development, and RA reverses the inhibited development caused by an angiogenesis inhibitor.


Assuntos
Inibidores da Angiogênese/farmacologia , Cinamatos/farmacologia , Rim/efeitos dos fármacos , Rim/crescimento & desenvolvimento , Tretinoína/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Biópsia por Agulha , Peso Corporal , Feminino , Imuno-Histoquímica , Córtex Renal/efeitos dos fármacos , Córtex Renal/crescimento & desenvolvimento , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Tamanho do Órgão/efeitos dos fármacos , Distribuição Aleatória , Estatísticas não Paramétricas
20.
Kidney Int ; 76(12): 1225-38, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19847153

RESUMO

Although the normal glomerulus comprises four resident cell types, least is known about the parietal epithelial cells (PECs). This comprehensive review addresses the cellular origin of PECs, discusses the normal structure and protein makeup of PECs, describes PEC function, and defines the responses to injury in disease and how these events lead to clinical events. The data show that PECs have unique properties and that new functions are being recognized such as their role in differentiating into podocytes during disease.


Assuntos
Glomérulos Renais/citologia , Animais , Apoptose , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Nefropatias/etiologia , Nefropatias/patologia , Nefropatias/fisiopatologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo , Modelos Biológicos , Permeabilidade , Podócitos/citologia , Podócitos/metabolismo , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA