Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Cells Tissues Organs ; 207(2): 69-82, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31671422

RESUMO

Clinical aspects dealing with the impairment of nephrogenesis in preterm and low birth weight babies were intensely researched. In this context it was shown that quite different noxae can harm nephron formation, and that the morphological damage in the fetal kidney is rather complex. Some pathological findings show that the impairment leads to changes in developing glomeruli that are restricted to the maturation zone of the outer cortex in the fetal human kidney. Other data show also imprints on the stages of nephron anlage including the niche, the pretubular aggregate, the renal vesicle, and comma- and S-shaped bodies located in the overlying nephrogenic zone of the rodent and human kidneys. During our investigations it was noticed that the stages of nephron anlage in the fetal human kidney during the phase of late gestation have not been described in detail. To contribute, these stages were recorded along with corresponding images. The initial nephron formation in the rodent kidney served as a reference. Finally, the known imprints left by the impairment in both specimens were listed and discussed. In sum, the relatively paucity of data on nephron formation in the fetal human kidney during the late phase of gestation is a call to start with intense research so that concepts for a therapeutic prolongation of nephrogenesis can be designed.


Assuntos
Feto/anormalidades , Rim , Néfrons , Organogênese , Animais , Feminino , Humanos , Recém-Nascido , Rim/anormalidades , Rim/anatomia & histologia , Rim/embriologia , Modelos Animais , Néfrons/anormalidades , Néfrons/anatomia & histologia , Néfrons/embriologia , Gravidez , Roedores
2.
Hipertens Riesgo Vasc ; 35(1): 24-29, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28431922

RESUMO

Renal macrocirculation participates in the development of arterial hypertension. The elevation in systemic blood pressure (BP) can damage the kidney starting in the microcirculation. Established arterial hypertension impinge upon the large arteries and stiffness develops. As a consequence central BP raises and BP pulsatility appear and contribute to further damage renal microcirculation by direct transmission of the elevated BP.


Assuntos
Hipertensão/patologia , Rim/patologia , Animais , Anti-Hipertensivos/uso terapêutico , Pressão Sanguínea/fisiologia , Cardiomegalia/etiologia , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Rim/irrigação sanguínea , Rim/fisiopatologia , Nefropatias/etiologia , Nefropatias/fisiopatologia , Microcirculação , Modelos Biológicos , Néfrons/anormalidades , Fluxo Pulsátil , Ratos , Ratos Endogâmicos SHR
3.
Am J Physiol Renal Physiol ; 313(3): F576-F584, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490528

RESUMO

Recently, new methods for assessing renal function in conscious mice (transcutaneous assessment) and for counting and sizing all glomeruli in whole kidneys (MRI) have been described. In the present study, these methods were used to assess renal structure and function in aging mice, and in mice born with a congenital low-nephron endowment. Age-related nephron loss was analyzed in adult C57BL/6 mice (10-50 wk of age), and congenital nephron deficit was assessed in glial cell line-derived neurotrophic factor heterozygous (GDNF HET)-null mutant mice. Renal function was measured through the transcutaneous quantitation of fluorescein isothiocyanate-sinistrin half-life (t1/2) in conscious mice. MRI was used to image, count, and size cationic-ferritin labeled glomeruli in whole kidneys ex vivo. Design-based stereology was used to validate the MRI measurements of glomerular number and mean volume. In adult C57BL/6 mice, older age was associated with fewer and larger glomeruli, and a rightward shift in the glomerular size distribution. These changes coincided with a decrease in renal function. GNDF HET mice had a congenital nephron deficit that was associated with glomerular hypertrophy and exacerbated by aging. These findings suggest that glomerular hypertrophy and hyperfiltration are compensatory processes that can occur in conjunction with both age-related nephron loss and congenital nephron deficiency. The combination of measurement of renal function in conscious animals and quantitation of glomerular number, volume, and volume distribution provides a powerful new tool for investigating aspects of renal aging and functional changes.


Assuntos
Envelhecimento/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Nefropatias/patologia , Nefropatias/fisiopatologia , Testes de Função Renal , Glomérulos Renais/patologia , Imageamento por Ressonância Magnética , Néfrons/anormalidades , Fatores Etários , Animais , Modelos Animais de Doenças , Fluoresceínas/administração & dosagem , Fluoresceínas/farmacocinética , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/farmacocinética , Predisposição Genética para Doença , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Taxa de Filtração Glomerular , Meia-Vida , Heterozigoto , Hipertrofia , Nefropatias/congênito , Glomérulos Renais/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligossacarídeos/administração & dosagem , Oligossacarídeos/farmacocinética , Fenótipo , Valor Preditivo dos Testes
4.
Dev Biol ; 427(1): 121-130, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28478097

RESUMO

In kidney development, connection of the nephric duct (ND) to the cloaca and subsequent sprouting of the ureteric bud (UB) from the ND are important for urinary exit tract formation. Although the roles of Ret signaling are well established, it remains unclear how intracellular cytoskeletal proteins regulate these morphogenetic processes. Myh9 and Myh10 encode two different non-muscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases. Here we report that ND/UB lineage-specific deletion of Myh9/Myh10 in mice caused severe hydroureter/hydronephrosis at birth. At mid-gestation, the mutant ND/UB epithelia exhibited aberrant basal protrusion and ectopic UB formation, which likely led to misconnection of the ureter to the bladder. In addition, the mutant epithelia exhibited apical extrusion followed by massive apoptosis in the lumen, which could be explained by reduced apical constriction and intercellular adhesion mediated by E-cadherin. These phenotypes were not ameliorated by genetic reduction of the tyrosine kinase receptor Ret. In contrast, ERK was activated in the mutant cells and its chemical inhibition partially ameliorated the phenotypes. Thus, myosin II is essential for maintaining the apicobasal integrity of the developing kidney epithelia independently of Ret signaling.


Assuntos
Epitélio/anormalidades , Rim/embriologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Ureter/anormalidades , Bexiga Urinária/anormalidades , Animais , Animais Recém-Nascidos , Cães , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Rim/metabolismo , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Néfrons/anormalidades , Néfrons/metabolismo , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Ureter/metabolismo , Bexiga Urinária/metabolismo
5.
PLoS One ; 11(7): e0158977, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27420727

RESUMO

During nephrogenesis, POU domain class 3 transcription factor 3 (POU3F3 aka BRN1) is critically involved in development of distinct nephron segments, including the thick ascending limb of the loop of Henle (TAL). Deficiency of POU3F3 in knock-out mice leads to underdevelopment of the TAL, lack of differentiation of TAL cells, and perinatal death due to renal failure. Pou3f3L423P mutant mice, which were established in the Munich ENU Mouse Mutagenesis Project, carry a recessive point mutation in the homeobox domain of POU3F3. Homozygous Pou3f3L423P mutants are viable and fertile. The present study used functional, as well as qualitative and quantitative morphological analyses to characterize the renal phenotype of juvenile (12 days) and aged (60 weeks) homo- and heterozygous Pou3f3L423P mutant mice and age-matched wild-type controls. In both age groups, homozygous mutants vs. control mice displayed significantly smaller kidney volumes, decreased nephron numbers and mean glomerular volumes, smaller TAL volumes, as well as lower volume densities of the TAL in the kidney. No histological or ultrastructural lesions of TAL cells or glomerular cells were observed in homozygous mutant mice. Aged homozygous mutants displayed increased serum urea concentrations and reduced specific urine gravity, but no evidence of glomerular dysfunction. These results confirm the role of POU3F3 in development and function of the TAL and provide new evidence for its involvement in regulation of the nephron number in the kidney. Therefore, Pou3f3L423P mutant mice represent a valuable research model for further analyses of POU3F3 functions, or for nephrological studies examining the role of congenital low nephron numbers.


Assuntos
Rim/anormalidades , Rim/crescimento & desenvolvimento , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Fatores do Domínio POU/genética , Animais , Pressão Sanguínea , Peso Corporal , Feminino , Rim/metabolismo , Alça do Néfron/anormalidades , Alça do Néfron/crescimento & desenvolvimento , Alça do Néfron/metabolismo , Masculino , Camundongos , Néfrons/anormalidades , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Tamanho do Órgão , RNA Mensageiro/genética
6.
Hum Mol Genet ; 25(3): 437-47, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26604140

RESUMO

Pallister-Hall syndrome (PHS) is a rare disorder caused by mutations in GLI3 that produce a transcriptional repressor (GLI3R). Individuals with PHS present with a variably penetrant variety of urogenital system malformations, including renal aplasia or hypoplasia, hydroureter, hydronephrosis or a common urogenital sinus. The embryologic mechanisms controlled by GLI3R that result in these pathologic phenotypes are undefined. We demonstrate that germline expression of GLI3R causes renal hypoplasia, associated with decreased nephron number, and hydroureter and hydronephrosis, caused by blind-ending ureters. Mice with obligate GLI3R expression also displayed duplication of the ureters that was caused by aberrant common nephric duct patterning and ureteric stalk outgrowth. These developmental abnormalities are associated with suppressed Hedgehog signaling activity in the cloaca and adjacent vesicular mesenchyme. Mice with conditional expression of GLI3R were utilized to identify lineage-specific effects of GLI3R. In the ureteric bud, GLI3R expression decreased branching morphogenesis. In Six2-positive nephrogenic progenitors, GLI3R decreased progenitor cell proliferation reducing the number of nephrogenic precursor structures. Using mutant mice with Gli3R and Gli3 null alleles, we demonstrate that urogenital system patterning and development is controlled by the levels of GLI3R and not by an absence of full-length GLI3. We conclude that the urogenital system phenotypes observed in PHS are caused by GLI3R-dependent perturbations in nephric duct patterning, renal branching morphogenesis and nephrogenic progenitor self-renewal.


Assuntos
Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Hidronefrose/genética , Rim/anormalidades , Fatores de Transcrição Kruppel-Like/genética , Proteínas do Tecido Nervoso/genética , Síndrome de Pallister-Hall/genética , Anormalidades Urogenitais/genética , Animais , Padronização Corporal/genética , Proliferação de Células , Modelos Animais de Doenças , Embrião de Mamíferos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hidronefrose/metabolismo , Hidronefrose/patologia , Rim/metabolismo , Rim/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Knockout , Mutação , Néfrons/anormalidades , Néfrons/embriologia , Néfrons/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Síndrome de Pallister-Hall/metabolismo , Síndrome de Pallister-Hall/patologia , Fenótipo , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ureter/anormalidades , Ureter/embriologia , Ureter/metabolismo , Anormalidades Urogenitais/metabolismo , Anormalidades Urogenitais/patologia , Proteína Gli3 com Dedos de Zinco
7.
Gene ; 572(1): 72-78, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26142106

RESUMO

Low nephron number has been recognised as an important cardiovascular risk factor and recently a strong correlation between renal mass and nephron number has been demonstrated in newborns. The aim of this study was to investigate individual, as well as combined, effects of common variants of genes which encode for major components of the renin-angiotensin system (REN G10601A, AGT G(-6)A, ACE I/D, AGTR1 A1166C) on kidney size in healthy, full-term newborns. A significant additive main effect of the ACE I/D polymorphism, as well as an additive-by-additive interaction between AGT G(-6)A and AGTR1 A1166C variants, were found. The variance attributed to the epistatic effect was 27.9 ml(2)/m(4), which accounted for 73.8% of the interaction variance (37.8 ml(2)/m(4)), 66.4% of the genetic variance (42.0 ml(2)/m(4)) and 4.4% to the total phenotypic variance (628 ml(2)/m(4)). No other statistically significant main or epistatic effects were detected. Our results highlight the importance of considering gene-gene interactions as part of the genetic architecture of congenital nephron number, even when the loci do not show significant single locus effects. Unravelling the genetic determinants of low nephron number, along with early molecular screening, may well help to identify children at risk for cardiovascular disease.


Assuntos
Angiotensinogênio/genética , Epistasia Genética , Rim/anatomia & histologia , Tamanho do Órgão/genética , Receptor Tipo 1 de Angiotensina/genética , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Feminino , Predisposição Genética para Doença , Variação Genética , Humanos , Recém-Nascido , Rim/anormalidades , Masculino , Modelos Genéticos , Néfrons/anormalidades , Néfrons/anatomia & histologia , Fenótipo , Polimorfismo de Nucleotídeo Único , Característica Quantitativa Herdável , Fatores de Risco
8.
Development ; 142(7): 1254-66, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25804736

RESUMO

Development of the metanephric kidney depends on tightly regulated interplay between self-renewal and differentiation of a nephron progenitor cell (NPC) pool. Several key factors required for the survival of NPCs have been identified, including fibroblast growth factor (FGF) signaling and the transcription factor Wilms' tumor suppressor 1 (WT1). Here, we present evidence that WT1 modulates FGF signaling by activating the expression of growth arrest-specific 1 (Gas1), a novel WT1 target gene and novel modulator of FGF signaling. We show that WT1 directly binds to a conserved DNA binding motif within the Gas1 promoter and activates Gas1 mRNA transcription in NPCs. We confirm that WT1 is required for Gas1 expression in kidneys in vivo. Loss of function of GAS1 in vivo results in hypoplastic kidneys with reduced nephron mass due to premature depletion of NPCs. Although kidney development in Gas1 knockout mice progresses normally until E15.5, NPCs show decreased rates of proliferation at this stage and are depleted as of E17.5. Lastly, we show that Gas1 is selectively required for FGF-stimulated AKT signaling in vitro. In summary, our data suggest a model in which WT1 modulates receptor tyrosine kinase signaling in NPCs by directing the expression of Gas1.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Néfrons/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas WT1/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células , DNA/genética , Ativação Enzimática/efeitos dos fármacos , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos Knockout , Modelos Animais , Néfrons/anormalidades , Néfrons/embriologia , Néfrons/patologia , Técnicas de Cultura de Órgãos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
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
10.
Am J Physiol Renal Physiol ; 306(7): F764-72, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24500693

RESUMO

MicroRNAs (miRNAs) are small, noncoding regulatory RNAs that act as posttranscriptional repressors by binding to the 3'-untranslated region (3'-UTR) of target genes. They require processing by Dicer, an RNase III enzyme, to become mature regulatory RNAs. Previous work from our laboratory revealed critical roles for miRNAs in nephron progenitors at midgestation (Ho J, Pandey P, Schatton T, Sims-Lucas S, Khalid M, Frank MH, Hartwig S, Kreidberg JA. J Am Soc Nephrol 22: 1053-1063, 2011). To interrogate roles for miRNAs in the early metanephric mesenchyme, which gives rise to nephron progenitors as well as the renal stroma during kidney development, we conditionally ablated Dicer function in this lineage. Despite normal ureteric bud outgrowth and condensation of the metanephric mesenchyme to form nephron progenitors, early loss of miRNAs in the metanephric mesenchyme resulted in severe renal dysgenesis. Nephron progenitors are initially correctly specified in the mutant kidneys, with normal expression of several transcription factors known to be critical in progenitors, including Six2, Pax2, Sall1, and Wt1. However, there is premature loss of the nephron progenitor marker Cited1, marked apoptosis, and increased expression of the proapoptotic protein Bim shortly after the initial inductive events in early kidney development. Subsequently, there is a failure in ureteric bud branching and nephron progenitor differentiation. Taken together, our data demonstrate a previously undetermined requirement for miRNAs during early kidney organogenesis and indicate a crucial role for miRNAs in regulating the survival of this lineage.


Assuntos
RNA Helicases DEAD-box/metabolismo , Células-Tronco Embrionárias/enzimologia , Rim/enzimologia , Mesoderma/enzimologia , Ribonuclease III/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Idade Gestacional , Rim/anormalidades , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesoderma/anormalidades , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Néfrons/anormalidades , Néfrons/enzimologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organogênese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ribonuclease III/deficiência , Ribonuclease III/genética , Transativadores/genética , Transativadores/metabolismo , Ureter/anormalidades , Ureter/enzimologia
11.
Reprod Fertil Dev ; 26(7): 1032-43, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23910917

RESUMO

A developmental insult that restricts growth in the first generation has the potential to program disease in subsequent generations. The aim of this study was to ascertain transgenerational growth and cardio-renal effects, via the maternal line, in a rat model of utero-placental insufficiency. Bilateral uterine vessel ligation or sham surgery (offspring termed first generation; F1 Restricted and Control, respectively) was performed in WKY rats. F1 Restricted and Control females were mated with normal males to produce second generation (F2) offspring (Restricted and Control) studied from fetal (embryonic Day 20) to adult (12 months) life. F2 Restricted male and female fetuses had reduced (P<0.05) nephron number (down 15-22%) but this deficit was not sustained postnatally and levels were similar to Controls at Day 35. F2 Restricted males, but not females, developed elevated (+16mmHg, P<0.05) systolic blood pressure at 6 months of age, which was sustained to 9 months. This was not explained by alterations to intra-renal or plasma components of the renin-angiotensin system. In a rat model of utero-placental insufficiency, we report alterations to F2 kidney development and sex-specific adult hypertension. This study demonstrates that low birthweight can have far-reaching effects that extend into the next generation.


Assuntos
Hipertensão/etiologia , Néfrons/anormalidades , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Peso Corporal , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Rim/embriologia , Masculino , Néfrons/embriologia , Tamanho do Órgão , Circulação Placentária/fisiologia , Insuficiência Placentária/fisiopatologia , Gravidez , Ratos , Ratos Endogâmicos WKY , Sistema Renina-Angiotensina , Fatores Sexuais , Útero/irrigação sanguínea
12.
Pediatrics ; 131(6): 1168-79, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23669525

RESUMO

Thanks to remarkable advances in neonatal intensive care, infants who once had little chance for survival can now enter adulthood. Yet the consequences of premature birth or low birth weight (LBW) on nephrogenesis, final nephron number, and long-term kidney function are unclear. This review focuses on the theory, experimental evidence, and observational data that suggest an increased risk of chronic kidney disease (CKD) for infants born prematurely. Many premature and LBW infants begin life with an incomplete complement of immature nephrons. They are then exposed to a variety of external stressors that can hinder ongoing kidney development or cause additional nephron loss such as hemodynamic alterations, nephrotoxic medications, infections, and suboptimal nutrition. Acute kidney injury, in particular, may be a significant risk factor for the development of CKD. According to Brenner's hypothesis, patients with decreased nephron number develop hyperfiltration that results in sodium retention, hypertension, nephron loss, and CKD due to secondary focal segmental glomerulosclerosis. Because the risk of CKD in premature and LBW infants has not been accurately determined, there are no evidence-based recommendations for screening or management. Yet with the first generation of infants from the surfactant era only now reaching adulthood, it is possible that there is already an unrecognized epidemic of CKD. We suggest individualized, risk-based assessments of premature and LBW infants due to the increased risk of CKD and call for additional research into the long-term risk for CKD these infants face.


Assuntos
Recém-Nascido Prematuro , Insuficiência Renal Crônica/epidemiologia , Adulto , Animais , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Testes de Função Renal , Néfrons/anormalidades , Medição de Risco , Fatores de Risco
13.
J Am Soc Nephrol ; 24(7): 1127-38, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23661808

RESUMO

DLG1 (discs-large homolog 1) and CASK (calcium/calmodulin-dependent serine protein kinase) interact at membrane-cytoskeleton interfaces and function as scaffolding proteins that link signaling molecules, receptors, and other scaffolding proteins at intercellular and synaptic junctions. Dlg1-null mice exhibit hydronephrosis, hydroureter, and occasionally hypoplastic kidneys, whereas Cask-null mice do not. To investigate whether DLG1 and CASK cooperate in the developing urogenital system, we generated mice deficient in both DLG1 and CASK either 1) globally, 2) in metanephric mesenchyme, or 3) in nephron progenitors. With each approach, Dlg1;Cask double-knockout (DKO) kidneys were severely hypoplastic and dysplastic and demonstrated rapid, premature depletion of nephron progenitors/stem cells. Several cellular and molecular defects were observed in the DKO kidneys, including reduced proliferation and increased apoptosis of cells in the nephrogenic zone and a progressive decrease in the number of cells expressing SIX2, a transcription factor essential for maintaining nephron progenitors. Fgf8 expression was reduced in early-stage DKO metanephric mesenchyme, accompanied by reduced levels of components of the Ras pathway, which is activated by fibroblast growth factor (FGF) signaling. Moreover, Dlg1(+/-);Cask(-/-) (het/null) kidneys were moderately hypoplastic and demonstrated impaired aggregation of SIX2-positive cells around the ureteric bud tips. Nephron progenitor-specific het/null mice survived with small kidneys but developed glomerulocystic kidney disease and renal failure. Taken together, these results suggest that DLG1 and CASK play critical cooperative roles in maintaining the nephron progenitor population, potentially via a mechanism involving effects on FGF signaling.


Assuntos
Diferenciação Celular , Guanilato Quinases/metabolismo , Néfrons/embriologia , Proteínas do Tecido Nervoso/metabolismo , Organogênese , Células-Tronco/citologia , Animais , Proteína 1 Homóloga a Discs-Large , Expressão Gênica , Camundongos , Camundongos Knockout , Néfrons/anormalidades , Néfrons/metabolismo , Proteínas Associadas SAP90-PSD95 , Transdução de Sinais
14.
Development ; 140(4): 886-96, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23362349

RESUMO

Nephron morphogenesis is a complex process that generates blood-filtration units (glomeruli) connected to extremely long and patterned tubular structures. Hepatocyte nuclear factor 1ß (HNF1ß) is a divergent homeobox transcription factor that is expressed in kidney from the first steps of nephrogenesis. Mutations in HNF1B (OMIM #137920) are frequently found in patients with developmental renal pathologies, the mechanisms of which have not been completely elucidated. Here we show that inactivation of Hnf1b in the murine metanephric mesenchyme leads to a drastic tubular defect characterized by the absence of proximal, distal and Henle's loop segments. Nephrons were eventually characterized by glomeruli, with a dilated urinary space, directly connected to collecting ducts via a primitive and short tubule. In the absence of HNF1ß early nephron precursors gave rise to deformed S-shaped bodies characterized by the absence of the typical bulge of epithelial cells at the bend between the mid and lower segments. The lack of this bulge eventually led to the absence of proximal tubules and Henle's loops. The expression of several genes, including Irx1, Osr2 and Pou3f3, was downregulated in the S-shaped bodies. We also observed decreased expression of Dll1 and the consequent defective activation of Notch in the prospective tubular compartment of comma- and S-shaped bodies. Our results reveal a novel hierarchical relationship between HNF1ß and key genes involved in renal development. In addition, these studies define a novel structural and functional component of S-shaped bodies at the origin of tubule formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 1-beta Nuclear de Hepatócito/metabolismo , Néfrons/embriologia , Organogênese/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator 1-beta Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Microscopia Eletrônica , Néfrons/anormalidades , Néfrons/ultraestrutura , Proteínas do Tecido Nervoso/metabolismo , Organogênese/genética , Fatores do Domínio POU/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/metabolismo
15.
J Perinat Med ; 41(3): 295-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23241663

RESUMO

AIMS: The study set out to investigate whether the osteopontin (OPN)-CD44-integrin-receptor-system is differently regulated during nephrogenesis in inborn nephron deficit, a major determinant of human primary hypertension and cardiovascular disease in adult life. METHODS: We compared a genetic rat model with an inherited nephron deficit, the Munich-Wistar-Froemter rat (MWF), to normotensive Wistar rats during nephrogenesis at day 19 of fetal development (E19) and at postpartal day 7 (D7). RESULTS: Renal OPN mRNA (-75%, P<0.05) and protein expression (-38%, P<0.05) were strongly decreased at E19 in MWF compared to Wistar. Renal mRNA-expression of CD44 was increased at E19 in MWF (+271%, P<0.05). At D7, renal OPN protein expression was increased (+115%, P<0.05) and renal mRNA-expression of CD44 remained elevated compared to Wistar control (+127%, P<0.05). CONCLUSIONS: Altered fetal expression of the OPN-CD44-integrin-receptor-system in the MWF model points to a possible role in low nephron number hypertension and cardiovascular disease.


Assuntos
Receptores de Hialuronatos/genética , Hipertensão Renal/genética , Hipertensão Renal/fisiopatologia , Néfrons/anormalidades , Osteopontina/genética , Animais , Modelos Animais de Doenças , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Integrina alfaV/genética , Néfrons/crescimento & desenvolvimento , Néfrons/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Mutantes , Ratos Wistar
16.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R682-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21653879

RESUMO

Suboptimal kidney development resulting from a genetic deficit in nephron number can have lifelong consequences that may lead to cardiorenal complications upon exposure to secondary insults in later life. To determine whether the inherited reduced renal reserve compromises the ability to handle osmotic stress in the adult animal, we challenged the heterozygous 3H1 Brachyrrhine (Br/+) mouse, which displays heritable renal hypoplasia associated with reduced embryonic six2 expression, to a solution of 2% NaCl for 5 days or to fluid restriction for 48 h. Blood chemistry, fluid intake, and physiological parameters, including renal measurements, were determined. Systemic hypertonicity by prolonged salt loading led to significant increases in plasma osmolality and plasma Na(+), along with polydipsia and polyuria, with a significant urine-concentrating defect that was resistant to DDAVP treatment in the adult Br/+ mouse compared with wild-type littermates. The Br/+ mouse also developed a significant increase in blood urea nitrogen at baseline that was further elevated when 2% NaCl was given. Fluid restriction for 48 h further enhanced plasma osmolality and plasma Na(+) responses, although the Br/+ mouse was evidently able to produce a small amount of concentrated urine at this time. Hypothalamic c-Fos expression was appropriately activated in the Br/+ mouse in response to both osmotic challenges, indicating an intact central neuroendocrine pathway that was not affected by the lack of congenital six2 expression. Collectively, our results demonstrate impaired osmoregulatory mechanisms consistent with chronic renal failure in the Br/+ mouse and indicate that six2 haploinsufficiency has a direct effect on postnatal fluid and electrolyte handling associated with fluid imbalance.


Assuntos
Falência Renal Crônica/metabolismo , Néfrons/metabolismo , Fatores de Transcrição/deficiência , Equilíbrio Hidroeletrolítico , Análise de Variância , Animais , Antidiuréticos/administração & dosagem , Nitrogênio da Ureia Sanguínea , Desamino Arginina Vasopressina/administração & dosagem , Ingestão de Líquidos , Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Proteínas de Homeodomínio/genética , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Capacidade de Concentração Renal , Falência Renal Crônica/genética , Falência Renal Crônica/fisiopatologia , Camundongos , Camundongos Mutantes , Néfrons/anormalidades , Néfrons/efeitos dos fármacos , Néfrons/fisiopatologia , Organogênese , Concentração Osmolar , Poliúria/genética , Poliúria/metabolismo , Poliúria/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Solução Salina Hipertônica/administração & dosagem , Solução Salina Hipertônica/metabolismo , Sódio/sangue , Cloreto de Sódio na Dieta/administração & dosagem , Cloreto de Sódio na Dieta/metabolismo , Fatores de Transcrição/genética , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/genética
17.
Rev. méd. hondur ; 79(2): 79-80, abr.-jun. 2011. ilus
Artigo em Espanhol | LILACS | ID: lil-644956

RESUMO

Introducción. La agenesia renal bilateral es una anomalía congénita mortal rara, originada por la ausencia del desarrollo de las nefronas y fallo de la división del esbozo ureteral. Caso clínico. Paciente femenino de 25 años de edad con embarazo de 33 semanas 3 días de gestación referida para nuevos estudios de imagen por su médico tratante debido a sospecha de agenesia renal bilateral en su feto, en ultrasonido convencional. No había historia personal o familiar relevante. Se le practicó un ultrasonido obstétrico convencional y doppler color con reconstrucciones tridimensionales, demostrando en el feto, la ausencia de las arterias renales y riñones, con signo de la suprarrenal tumbada. Se observó, además agenesia vesical, mesocardia con dextroposición, tórax en campana con hipoplasia pulmonar y anhidramnios. Ocurrió óbito a las 35 semanas. Conclusión. Los hallazgos fueron congruentes con agenesia renal bilateral, con pronóstico fatal en el tercer trimestre del embarazo.


Assuntos
Humanos , Adulto , Feminino , Complicações na Gravidez , Néfrons/anormalidades , Rim/anormalidades , Transmissão Vertical de Doenças Infecciosas , Ultrassonografia Doppler/métodos
18.
Rev. méd. hondur ; 79(2): 79-80, abr.-jun. 2011. ilus
Artigo em Espanhol | BIMENA | ID: bim-5401

RESUMO

Introducción. La agenesia renal bilateral es una anomalía congénita mortal rara, originada por la ausencia del desarrollo de las nefronas y fallo de la división del esbozo ureteral. Caso clínico. Paciente femenino de 25 años de edad con embarazo de 33 semanas 3 días de gestación referida para nuevos estudios de imagen por su médico tratante debido a sospecha de agenesia renal bilateral en su feto, en ultrasonido convencional. No había historia personal o familiar relevante. Se le practicó un ultrasonido obstétrico convencional y doppler color con reconstrucciones tridimensionales, demostrando en el feto, la ausencia de las arterias renales y riñones, con signo de la suprarrenal tumbada. Se observó, además agenesia vesical, mesocardia con dextroposición, tórax en campana con hipoplasia pulmonar y anhidramnios. Ocurrió óbito a las 35 semanas. Conclusión. Los hallazgos fueron congruentes con agenesia renal bilateral, con pronóstico fatal en el tercer trimestre del embarazo.


Assuntos
Humanos , Feminino , Adulto , Rim/anormalidades , Néfrons/anormalidades , Complicações na Gravidez , Transmissão Vertical de Doenças Infecciosas , Ultrassonografia Doppler/métodos
19.
Pediatr Nephrol ; 26(9): 1453-61, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21327776

RESUMO

Congenital obstructive nephropathy is the leading cause of chronic renal disease in children. As a result, it represents a tremendous societal burden in terms of morbidity and mortality, as well as in health care expenses of caring for children with chronic kidney disease and end-stage renal disease. The various diagnostic, prognostic, and therapeutic challenges associated with congenital obstructive nephropathy highlight the importance of developing effective experimental models for studying this disease process. In this review, we define the clinical entity that is congenital obstructive nephropathy, outline the current standards of diagnosis and care, and discuss the utilization of current experimental models designed to help clarify some of the clinical conundrums associated with this important disease.


Assuntos
Rim/anormalidades , Néfrons/anormalidades , Anormalidades Urogenitais , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Rim/fisiopatologia , Néfrons/fisiopatologia , Prognóstico , Fatores de Risco , Anormalidades Urogenitais/complicações , Anormalidades Urogenitais/diagnóstico , Anormalidades Urogenitais/epidemiologia , Anormalidades Urogenitais/fisiopatologia , Anormalidades Urogenitais/terapia
20.
Am J Physiol Renal Physiol ; 300(1): F147-56, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962117

RESUMO

A hyperglycemic environment in utero reduces kidney size and nephron number due to nascent nephron apoptosis. However, the underlying mechanisms are incompletely understood. The present study investigated whether the nascent nephron apoptosis promoted by high glucose is mediated via the transcription factor NF-κB and p53 signaling pathways. Neonatal mouse kidneys from the offspring of nondiabetic, diabetic, and insulin-treated diabetic dams were used for in vivo studies, and MK4 cells, an embryonic metanephric mesenchymal (MM) cell line, were used for in vitro studies. Neonatal kidneys of the offspring of diabetic mothers exhibited an increased number of apoptotic cells and reactive oxygen species (ROS) generation, enhanced NF-κB activation, and nuclear translocation of its subunits (p50 and p65 subunits) as well as phosphorylation (Ser 15) of p53 compared with kidneys of offspring of nondiabetic mothers. Insulin treatment of diabetic dams normalized these parameters in the offspring. In vitro, high-glucose (25 mM) induced ROS generation and significantly increased MK4 cell apoptosis and caspase-3 activity via activation of NF-κB pathway, with p53 phosphorylation and nuclear translocation compared with normal glucose (5 mM). These changes in a high-glucose milieu were prevented by transient transfection of small interfering RNAs for dominant negative IκBα or IKK or p53. Our data demonstrate that high glucose-induced nascent nephron apoptosis is mediated, at least in part, via ROS generation and the activation of NF-κB and p53 pathways.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Experimental/fisiopatologia , Glucose/farmacologia , NF-kappa B/fisiologia , Néfrons/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Animais , Linhagem Celular , Feminino , Glucose/administração & dosagem , Insulina/uso terapêutico , Rim/anormalidades , Camundongos , Subunidade p50 de NF-kappa B/biossíntese , Néfrons/anormalidades , Gravidez , Gravidez em Diabéticas/tratamento farmacológico , Gravidez em Diabéticas/fisiopatologia , Fator de Transcrição RelA/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...