Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Semin Pediatr Surg ; 31(6): 151232, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36423515

RESUMO

Congenital anomalies of the kidney and urinary tract or "CAKUT" describes a spectrum of developmental disorders with a range of associated clinical presentations and functional consequences. CAKUT underlies the majority of chronic kidney disease and kidney replacement therapy requirement in children, but functional deterioration can also emerge in adulthood. Understanding the normal embryological processes involved in kidney development allows us to appreciate the timing and sequence of critical events implicated when things go wrong. In this review, we will describe the normal developmental mechanisms and relate this to what we currently know about the pathological processes involved in various forms of CAKUT. We will also review the proposed etiological factors, in particular genetics, involved in CAKUT.


Assuntos
Sistema Urinário , Anormalidades Urogenitais , Refluxo Vesicoureteral , Criança , Humanos , Adulto , Sistema Urinário/cirurgia , Sistema Urinário/anormalidades , Rim/cirurgia , Rim/anormalidades , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/cirurgia , Anormalidades Urogenitais/patologia
2.
Am J Med Genet C Semin Med Genet ; 190(3): 264-278, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36165461

RESUMO

Pallister-Hall syndrome (PHS) is a rare autosomal dominant disease diagnosed by the presence of hypothalamic hamartoma, mesoaxial polydactyly and a truncating variant in the middle third of the GLI-Kruppel family member 3 (GLI3) gene. PHS may also include a wide range of clinical phenotypes affecting multiple organ systems including congenital anomalies of the kidney and urinary tract (CAKUT). The observed clinical phenotypes are consistent with the essential role of GLI3, a transcriptional effector in the hedgehog (Hh) signaling pathway, in organogenesis. However, the mechanisms by which truncation of GLI3 in PHS results in such a variety of clinical phenotypes with variable severity, even within the same organ, remain unclear. In this study we focus on presentation of CAKUT in PHS. A systematic analysis of reported PHS patients (n = 78) revealed a prevalence of 26.9% (21/78) of CAKUT. Hypoplasia (± dysplasia) and agenesis were the two main types of CAKUT; bilateral and unilateral CAKUT were reported with equal frequency. Examination of clinical phenotypes with CAKUT revealed a significant association between CAKUT and craniofacial defects, bifid epiglottis and a Disorder of Sex Development, specifically affecting external genitalia. Lastly, we determined that PHS patients with CAKUT predominately had substitution type variants (as opposed to deletion type variants in non-CAKUT PHS patients) in the middle third of the GLI3 gene. These results provide a foundation for future work aimed at uncovering the molecular mechanisms by which variant GLI3 result in the wide range and severity of clinical features observed in PHS.


Assuntos
Anormalidades Múltiplas , Síndrome de Pallister-Hall , Sistema Urinário , Humanos , Síndrome de Pallister-Hall/diagnóstico , Síndrome de Pallister-Hall/genética , Proteína Gli3 com Dedos de Zinco/genética , Fatores de Transcrição Kruppel-Like/genética , Anormalidades Múltiplas/genética , Proteínas do Tecido Nervoso/genética , Proteínas Hedgehog , Rim
3.
Pediatrics ; 148(2)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34230093

RESUMO

BACKGROUND AND OBJECTIVES: Children with isolated unilateral multicystic dysplastic kidney (MCDK) or congenital solitary kidney (CSK) undergo serial renal ultrasonography with variable frequency until they are transitioned to adult care. A growing body of literature suggests the value of frequent ultrasonography in this population is limited, providing no benefit to overall outcomes. Despite emerging evidence, ultrasound remains overused, resulting in avoidable health care expenditures and unnecessary use of resources. With our initiative, we aimed to improve quality of care by reducing avoidable ultrasounds in these children. METHODS: This was a single-center, prospective, interrupted time series of children <18 years with ultrasound-confirmed isolated unilateral MCDK or CSK in the outpatient nephrology clinic to evaluate the effect of a decision-making algorithm on the proportion of children receiving an avoidable ultrasound. An algorithm depicting a consensus, evidence-based protocol for managing pediatric MCDK or CSK was refined through content expert feedback and usability testing to standardize frequency of ultrasonography. Ultrasounds were deemed necessary after birth, at 6 months, and at 2, 5, 10, and 15 years. Differences pre- and postintervention were determined by using a U chart and t and F tests for significance. RESULTS: The algorithm resulted in a 47% reduction (P < .001) in the proportion of avoidable ultrasounds ordered in children with MCDK and CSK. This reduction was sustainable over a 6-month period and would result in at least $46 000 annual savings. CONCLUSIONS: Introduction of a clinical decision-making algorithm was associated with a reduction in avoidable ultrasound testing. Improving adherence across providers may allow for an even more pronounced reduction.


Assuntos
Rim Displásico Multicístico/diagnóstico por imagem , Rim Único/diagnóstico por imagem , Procedimentos Desnecessários/estatística & dados numéricos , Adolescente , Algoritmos , Criança , Pré-Escolar , Humanos , Lactente , Análise de Séries Temporais Interrompida , Estudos Prospectivos , Ultrassonografia/estatística & dados numéricos
4.
Development ; 147(3)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31932349

RESUMO

Cerebellar granule cell (GC) development relies on precise regulation of sonic hedgehog (Shh)-Gli signalling activity, failure of which is associated with motor disorders and medulloblastoma. Mutations in the pathway regulator suppressor of fused (Sufu), which modulates Gli activators and repressors, are linked to cerebellar dysfunction and tumourigenesis. The mechanism by which Sufu calibrates Shh signalling in GCs is unknown. Math1-Cre-mediated deletion of Sufu in mouse GC progenitors (GCPs) demonstrated that Sufu restricts GCP proliferation and promotes cell cycle exit, by promoting expression of Gli3R and suppressing Gli2 levels. Sufu is also required to promote a high threshold of pathway activity in GCPs. Remarkably, central cerebellar lobules are more deleteriously impacted by Sufu deletion, but are less sensitive to downstream genetic manipulations to reduce Gli2 expression or overexpress a Gli3R mimic, compared with anterior lobules. Transcriptome sequencing uncovered new Sufu targets, especially Fgf8, which is upregulated in Sufu-mutant GCPs. We demonstrate that Fgf8 is necessary and sufficient to drive Sufu-mutant GCP proliferation. This study reveals new insights into the spatial and temporal regulation of cerebellar Shh-Gli signalling, while uncovering new targets, such as Fgf8.


Assuntos
Proliferação de Células/genética , Cerebelo/citologia , Fator 8 de Crescimento de Fibroblasto/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Proteína Gli3 com Dedos de Zinco/metabolismo , Animais , Ciclo Celular/genética , Cerebelo/crescimento & desenvolvimento , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Repressoras/genética , Transdução de Sinais/genética , Transcriptoma , Proteína Gli2 com Dedos de Zinco/genética , Proteína Gli3 com Dedos de Zinco/genética
5.
Pediatr Nephrol ; 35(5): 725-731, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-30923969

RESUMO

Aberrant hedgehog (Hh) signaling during embryogenesis results in various severe congenital abnormalities, including renal malformations. The molecular mechanisms that underlie congenital renal malformations remain poorly understood. Here, we review the current understanding of the lineage-specific roles of Hh signaling during renal morphogenesis and how aberrant Hh signaling during embryonic kidney development contributes to renal malformation.


Assuntos
Desenvolvimento Embrionário/genética , Proteínas Hedgehog/metabolismo , Rim/embriologia , Transdução de Sinais/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Animais , Linhagem da Célula/genética , Embrião de Mamíferos/anormalidades , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Humanos , Rim/anormalidades , Rim/citologia , Camundongos , Modelos Animais , Mutação , Proteína GLI1 em Dedos de Zinco/genética
6.
Development ; 145(13)2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29945868

RESUMO

Normal kidney function depends on the proper development of the nephron: the functional unit of the kidney. Reciprocal signaling interactions between the stroma and nephron progenitor compartment have been proposed to control nephron development. Here, we show that removal of hedgehog intracellular effector smoothened (Smo-deficient mutants) in the cortical stroma results in an abnormal renal capsule, and an expanded nephron progenitor domain with an accompanying decrease in nephron number via a block in epithelialization. We show that stromal-hedgehog-Smo signaling acts through a GLI3 repressor. Whole-kidney RNA sequencing and analysis of FACS-isolated stromal cells identified impaired TGFß2 signaling in Smo-deficient mutants. We show that neutralization and knockdown of TGFß2 in explants inhibited nephrogenesis. In addition, we demonstrate that concurrent deletion of Tgfbr2 in stromal and nephrogenic cells in vivo results in decreased nephron formation and an expanded nephrogenic precursor domain similar to that observed in Smo-deficient mutant mice. Together, our data suggest a mechanism whereby a stromal hedgehog-TGFß2 signaling axis acts to control nephrogenesis.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas Hedgehog/metabolismo , Néfrons/embriologia , Transdução de Sinais/fisiologia , Receptor Smoothened/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Fatores de Transcrição Forkhead/genética , Proteínas Hedgehog/genética , Camundongos , Camundongos Knockout , Néfrons/citologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptor Smoothened/genética , Células Estromais/citologia , Células Estromais/metabolismo , Fator de Crescimento Transformador beta2/genética , Proteína Gli3 com Dedos de Zinco/genética , Proteína Gli3 com Dedos de Zinco/metabolismo
7.
J Am Soc Nephrol ; 29(4): 1198-1209, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436516

RESUMO

Nonobstructive hydronephrosis, defined as dilatation of the renal pelvis with or without dilatation of the ureter, is the most common antenatal abnormality detected by fetal ultrasound. Yet, the etiology of nonobstructive hydronephrosis is poorly defined. We previously demonstrated that defective development of urinary tract pacemaker cells (utPMCs) expressing hyperpolarization-activated cyclic nucleotide-gated channel 3 (HCN3) and the stem cell marker cKIT causes abnormal ureteric peristalsis and nonobstructive hydronephrosis. However, further investigation of utPMC development and function is limited by lack of knowledge regarding the embryonic derivation, development, and molecular apparatus of these cells. Here, we used lineage tracing in mice to identify cells that give rise to utPMCs. Neural crest cells (NCCs) indelibly labeled with tdTomato expressed HCN3 and cKIT. Furthermore, purified HCN3+ and cKIT+ utPMCs were enriched in Sox10 and Tfap-2α, markers of NCCs. Sequencing of purified RNA from HCN3+ cells revealed enrichment of a small subset of RNAs, including RNA encoding protein kinase 2ß (PTK2ß), a Ca2+-dependent tyrosine kinase that regulates ion channel activity in neurons. Immunofluorescence analysis in situ revealed PTK2ß expression in NCCs as early as embryonic day 12.5 and in HCN3+ and cKIT+ utPMCs as early as embryonic day 15.5, with sustained expression in HCN3+ utPMCs until postnatal week 8. Pharmacologic inhibition of PTK2ß in murine pyeloureteral tissue explants inhibited contraction frequency. Together, these results demonstrate that utPMCs are derived from NCCs, identify new markers of utPMCs, and demonstrate a functional contribution of PTK2ß to utPMC function.


Assuntos
Quinase 2 de Adesão Focal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células Intersticiais de Cajal/enzimologia , Pelve Renal/fisiologia , Crista Neural/enzimologia , Peristaltismo/fisiologia , Ureter/fisiologia , Animais , Antígenos de Diferenciação/análise , Quinase 2 de Adesão Focal/biossíntese , Quinase 2 de Adesão Focal/genética , Genes Reporter , Idade Gestacional , Hidronefrose/enzimologia , Hidronefrose/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/análise , Células Intersticiais de Cajal/fisiologia , Pelve Renal/citologia , Pelve Renal/embriologia , Pelve Renal/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Crista Neural/fisiologia , Canais de Potássio/análise , Proteínas Proto-Oncogênicas c-kit/análise , RNA Mensageiro/biossíntese , Fatores de Transcrição SOXE/análise , Transdução de Sinais , Fator de Transcrição AP-2/análise , Ureter/citologia , Ureter/embriologia , Ureter/crescimento & desenvolvimento
8.
Dev Dyn ; 247(1): 156-169, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28560839

RESUMO

BACKGROUND: Deficiency of Suppressor of Fused (SuFu), an intracellular mediator of Hedgehog signaling, in the murine mid-hindbrain disrupts cerebellar morphogenesis and cell differentiation in a manner that is rescued by constitutive expression of GLI3 transcriptional repressor (GLI3R). Here, we determined SuFu functions in cerebellar radial precursors following the stage of mid-hindbrain specification using a Blbp-Cre transgene. RESULTS: SuFu-deficient cerebella were severely dysplastic, and characterized by laminar disorganization, and delayed differentiation of ventricular zone-derived precursors. In vitro analysis of cerebellar precursors isolated from control and mutant mice demonstrated an increased proportion of radial glial precursors vs. Tuj1-positive neurons in mutant cultures. Abnormal cell differentiation in SuFu-deficient precursors was rescued by a constitutively expressed GLI3R knock-in allele, albeit with variable penetrance. Using RNA expression analysis in control and SuFu-deficient cerebellar anlage, we identified up-regulation of Fgf15 in mutant tissue. Strikingly, exogenous hFGF19, a mFGF15 ortholog, inhibited neuronal differentiation in cultures of wild-type cerebellar precursors. Moreover, siRNA-mediated knockdown of Fgf15 in SuFu-deficient cerebellar precursors rescued their delayed differentiation to neurons. CONCLUSIONS: Together, our results show that SuFu promotes cerebellar radial precursor differentiation to neurons. SuFu function is mediated in part by GLI3R and down-regulation of Fgf15 expression. Developmental Dynamics 247:156-169, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular/fisiologia , Cerebelo/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Proteína Gli3 com Dedos de Zinco/metabolismo , Animais , Cerebelo/citologia , Regulação para Baixo , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Camundongos , Camundongos Transgênicos , Neurogênese/fisiologia , Neurônios/citologia , RNA Interferente Pequeno , Transdução de Sinais/fisiologia
9.
J Am Soc Nephrol ; 29(2): 532-544, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29109083

RESUMO

Intrinsic ureteropelvic junction obstruction is the most common cause of congenital hydronephrosis, yet the underlying pathogenesis is undefined. Hedgehog proteins control morphogenesis by promoting GLI-dependent transcriptional activation and inhibiting the formation of the GLI3 transcriptional repressor. Hedgehog regulates differentiation and proliferation of ureteric smooth muscle progenitor cells during murine kidney-ureter development. Histopathologic findings of smooth muscle cell hypertrophy and stroma-like cells, consistently observed in obstructing tissue at the time of surgical correction, suggest that Hedgehog signaling is abnormally regulated during the genesis of congenital intrinsic ureteropelvic junction obstruction. Here, we demonstrate that constitutively active Hedgehog signaling in murine intermediate mesoderm-derived renal progenitors results in hydronephrosis and failure to develop a patent pelvic-ureteric junction. Tissue obstructing the ureteropelvic junction was marked as early as E13.5 by an ectopic population of cells expressing Ptch2, a Hedgehog signaling target. Constitutive expression of GLI3 repressor in Ptch1-deficient mice rescued ectopic Ptch2 expression and obstructive hydronephrosis. Whole transcriptome analysis of isolated Ptch2+ cells revealed coexpression of genes characteristic of stromal progenitor cells. Genetic lineage tracing indicated that stromal cells blocking the ureteropelvic junction were derived from intermediate mesoderm-derived renal progenitors and were distinct from the smooth muscle or epithelial lineages. Analysis of obstructive ureteric tissue resected from children with congenital intrinsic ureteropelvic junction obstruction revealed a molecular signature similar to that observed in Ptch1-deficient mice. Together, these results demonstrate a Hedgehog-dependent mechanism underlying mammalian intrinsic ureteropelvic junction obstruction.


Assuntos
Proteínas Hedgehog/genética , Hidronefrose/genética , Proteínas do Tecido Nervoso/genética , Receptor Patched-1/genética , Receptor Patched-2/genética , Transdução de Sinais , Obstrução Ureteral/genética , Proteína Gli3 com Dedos de Zinco/genética , Aldeído Oxirredutases/genética , Animais , Linhagem da Célula , Criança , Feminino , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Hidronefrose/congênito , Hidronefrose/patologia , Hibridização In Situ , Pelve Renal/embriologia , Pelve Renal/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Transcriptoma , Regulação para Cima , Ureter/embriologia , Ureter/metabolismo , Obstrução Ureteral/congênito , Obstrução Ureteral/patologia , Proteína Gli3 com Dedos de Zinco/metabolismo
10.
J Am Soc Nephrol ; 27(5): 1465-77, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26407593

RESUMO

Integrin-linked kinase (ILK) is an intracellular scaffold protein with critical cell-specific functions in the embryonic and mature mammalian kidney. Previously, we demonstrated a requirement for Ilk during ureteric branching and cell cycle regulation in collecting duct cells in vivo Although in vitro data indicate that ILK controls p38 mitogen-activated protein kinase (p38MAPK) activity, the contribution of ILK-p38MAPK signaling to branching morphogenesis in vivo is not defined. Here, we identified genes that are regulated by Ilk in ureteric cells using a whole-genome expression analysis of whole-kidney mRNA in mice with Ilk deficiency in the ureteric cell lineage. Six genes with expression in ureteric tip cells, including Wnt11, were downregulated, whereas the expression of dual-specificity phosphatase 8 (DUSP8) was upregulated. Phosphorylation of p38MAPK was decreased in kidney tissue with Ilk deficiency, but no significant decrease in the phosphorylation of other intracellular effectors previously shown to control renal morphogenesis was observed. Pharmacologic inhibition of p38MAPK activity in murine inner medullary collecting duct 3 (mIMCD3) cells decreased expression of Wnt11, Krt23, and Slo4c1 DUSP8 overexpression in mIMCD3 cells significantly inhibited p38MAPK activation and the expression of Wnt11 and Slo4c1. Adenovirus-mediated overexpression of DUSP8 in cultured embryonic murine kidneys decreased ureteric branching and p38MAPK activation. Together, these data demonstrate that Ilk controls branching morphogenesis by regulating the expression of DUSP8, which inhibits p38MAPK activity and decreases branching morphogenesis.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Rim/embriologia , Rim/enzimologia , Morfogênese , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Camundongos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
11.
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
12.
Clin J Am Soc Nephrol ; 10(10): 1783-90, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26231195

RESUMO

BACKGROUND AND OBJECTIVES: Standard clinical assessments do not predict surgical intervention in patients with a moderate degree of upper tract hydronephrosis. This study investigated whether combined measures of renal calyceal dilation and anteroposterior diameter (APD) of the renal pelvis at the first postnatal ultrasound better predict surgical intervention beyond standard assessments of the APD or Society of Fetal Urology (SFU) grading system. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: A retrospective cohort of 348 children with antenatal hydronephrosis followed from 2003 to 2013 were studied. Using Cox regression, the risk for surgery by APD, SFU, and combined grading on the basis of the first postnatal ultrasound was calculated. The predictive capability of each grading system for surgery was determined by calculating the positive likelihood ratio (LR+). RESULTS: The combination of APD≥6-9 mm and diffuse caliectasis had a hazard ratio (HR) of 19.5 (95% confidence interval [95% CI], 3.94 to 96.9) versus 0.59 (95% CI, 0.05 to 6.53) for APD≥6-9 mm alone and a similar risk of 8.9 for SFU grade 3 (95% CI, 3.84 to 20.9). The combination of APD≥9-15 mm and diffuse caliectasis had an HR of 18.7 (95% CI, 4.36 to 80.4) versus 1.75 (95% CI, 0.29 to 10.5) for APD≥9-15 mm alone. The LR+ for surgery for diffuse caliectasis and APD≥6-9 mm was higher than for APD≥6-9 mm alone (HR=2.62; 95% CI, 0.87 to 7.94 versus HR=0.04; 95% CI, 0.01 to 0.32) and was higher for APD≥9-15 mm and diffuse caliectasis than APD≥9-15 mm alone (HR=2.0; 95% CI, 1.15 to 3.45 versus HR=0.14; 95% CI, 0.04 to 0.43). Both combined groups of moderate hydronephrosis (APD≥6-9 mm or ≥9-15 mm with diffuse caliectasis) had only slightly higher LR+ than SFU grade 3 (HR=1.89; 95% CI, 1.17 to 3.05). CONCLUSIONS: These results suggest a grading system combining APD and diffuse caliectasis distinguishes those children with moderate degrees of upper tract hydronephrosis that are at higher risk of surgery.


Assuntos
Hidronefrose/diagnóstico por imagem , Hidronefrose/cirurgia , Cálices Renais/diagnóstico por imagem , Índice de Gravidade de Doença , Dilatação Patológica/diagnóstico por imagem , Feminino , Humanos , Lactente , Recém-Nascido , Cálices Renais/patologia , Pelve Renal/diagnóstico por imagem , Funções Verossimilhança , Masculino , Cuidado Pós-Natal , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Curva ROC , Estudos Retrospectivos , Fatores de Risco , Ultrassonografia Pré-Natal , Obstrução Ureteral/complicações , Obstrução Ureteral/cirurgia
13.
J Histochem Cytochem ; 63(1): 32-44, 2015 01.
Artigo em Inglês | MEDLINE | ID: mdl-25319549

RESUMO

Schimke immuno-osseous dysplasia (SIOD) is a pleiotropic disorder caused by mutations in the SWI/SNF2-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like-1 (SMARCAL1) gene, with multiple clinical features, notably end-stage renal disease. Here we characterize the renal pathology in SIOD patients. Our analysis of SIOD patient renal biopsies demonstrates the tip and collapsing variants of focal segmental glomerulosclerosis (FSGS). Additionally, electron microscopy revealed numerous glomerular abnormalities most notably in the podocyte and Bowman's capsule. To better understand the role of SMARCAL1 in the pathogenesis of FSGS, we defined SMARCAL1 expression in the developing and mature kidney. In the developing fetal kidney, SMARCAL1 is expressed in the ureteric epithelium, stroma, metanephric mesenchyme, and in all stages of the developing nephron, including the maturing glomerulus. In postnatal kidneys, SMARCAL1 expression is localized to epithelial tubules of the nephron, collecting ducts, and glomerulus (podocytes and endothelial cells). Interestingly, not all cells within the same lineage expressed SMARCAL1. In renal biopsies from SIOD patients, TUNEL analysis detected marked increases in DNA fragmentation. Our results highlight the cells that may contribute to the renal pathogenesis in SIOD. Further, we suggest that disruptions in genomic integrity during fetal kidney development contribute to the pathogenesis of FSGS in SIOD patients.


Assuntos
Arteriosclerose/metabolismo , Arteriosclerose/patologia , DNA Helicases/metabolismo , Regulação da Expressão Gênica , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/patologia , Rim/metabolismo , Rim/patologia , Síndrome Nefrótica/metabolismo , Síndrome Nefrótica/patologia , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patologia , Animais , Arteriosclerose/complicações , Arteriosclerose/genética , Criança , Pré-Escolar , Fragmentação do DNA , Feminino , Glomerulosclerose Segmentar e Focal/complicações , Humanos , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/genética , Rim/embriologia , Rim/ultraestrutura , Masculino , Camundongos , Síndrome Nefrótica/complicações , Síndrome Nefrótica/genética , Osteocondrodisplasias/complicações , Osteocondrodisplasias/genética , Doenças da Imunodeficiência Primária , Embolia Pulmonar/complicações , Embolia Pulmonar/genética
14.
Am J Pathol ; 184(5): 1395-410, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24637293

RESUMO

Renal dysplasia, a developmental disorder characterized by defective ureteric branching morphogenesis and nephrogenesis, ranks as one of the major causes of renal failure among the pediatric population. Herein, we demonstrate that the levels of activated ß-catenin are elevated in the nuclei of ureteric, stromal, and mesenchymal cells within dysplastic human kidney tissue. By using a conditional mouse model of mesenchymal ß-catenin overexpression, we identify two novel signaling pathways mediated by ß-catenin in the development of renal dysplasia. First, the overexpression of ß-catenin within the metanephric mesenchyme leads to ectopic and disorganized branching morphogenesis caused by ß-catenin directly binding Tcf/lef consensus binding sites in the Gdnf promoter and up-regulating Gdnf transcription. Second, ß-catenin overexpression in the metanephric mesenchyme leads to elevated levels of transcriptionally active ß-catenin in the ureteric epithelium. Interestingly, this increase of ß-catenin-mediated transcription results from a novel Ret/ß-catenin signaling pathway. Consistent with these findings, analysis of human dysplastic renal tissue demonstrates that undifferentiated mesenchymal cells expressing high levels of ß-catenin also express increased GDNF. Furthermore, dysplastic ureteric tubules that were surrounded by high levels of GDNF also exhibited increased levels of activated ß-catenin. Together, these data support a model in which the elevation of ß-catenin in the metanephric mesenchyme results in cell-autonomous and non-cell-autonomous events that lead to the genesis of renal dysplasia.


Assuntos
Nefropatias/patologia , Rim/anormalidades , Rim/patologia , Mesoderma/metabolismo , Mesoderma/patologia , beta Catenina/metabolismo , Animais , Feminino , Regulação da Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células HEK293 , Humanos , Rim/embriologia , Nefropatias/metabolismo , Masculino , Camundongos , Modelos Biológicos , Morfogênese , Mutação , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas Wnt/metabolismo
15.
PLoS One ; 8(6): e65448, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762375

RESUMO

The primary cilium is required during early embryo patterning, epithelial tubulogenesis, and growth factor-dependent signal transduction. The requirement for primary cilia during renal epithelial-mesenchymal tissue interactions that give rise to nephrons is undefined. Here, we used Cre-mediated recombination to generate mice with Kif3a deficiency targeted to the ureteric and/or metanephric mesenchyme cell lineages in the embryonic kidney. Gradual loss of primary cilia in either lineage leads to a phenotype of reduced nephron number. Remarkably, in addition to cyst formation, loss of primary cilia in the ureteric epithelial cell leads to decreased expression of Wnt11 and Ret and reduced ureteric branching. Constitutive expression of GLI3 repressor (Gli3(Δ699/+) ) rescues these abnormalities. In embryonic metanephric mesenchyme cells, Kif3a deficiency limits survival of nephrogenic progenitor cells and expression of genes required for nephron formation. Together, our data demonstrate that Kif3a controls nephron number via distinct cell lineage-specific mechanisms.


Assuntos
Linhagem da Célula , Regulação da Expressão Gênica , Cinesinas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Néfrons/citologia , Néfrons/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cílios/efeitos dos fármacos , Cílios/metabolismo , Cílios/ultraestrutura , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Cinesinas/deficiência , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Néfrons/crescimento & desenvolvimento , Néfrons/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tamoxifeno/farmacologia , Proteína Gli3 com Dedos de Zinco
16.
Nat Rev Nephrol ; 9(7): 409-18, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23752887

RESUMO

Alagille syndrome is an autosomal dominant disorder with variable multisystem organ involvement that is caused by mutations in one of two genes in the Notch signalling pathway, JAG1 or NOTCH2. Alagille syndrome is characterized by bile duct paucity, along with at least three of the following features: cholestasis, cardiac defects, skeletal abnormalities, ocular abnormalities and characteristic facies. However, the clinical features of Alagille syndrome are highly variable, and children or adults may also present with predominantly renal findings and little or no hepatic involvement. Renal involvement occurs in 40% of JAG1-mutation-positive individuals. Renal insufficiency is common and has been specifically reported in children with Alagille syndrome who have end-stage liver disease. The role of NOTCH2 and JAG1 in formation of proximal nephron structures and podocytes might explain the observed phenotypes of renal dysplasia and proteinuria in patients with Alagille syndrome, and renal tubular acidosis may be the result of JAG1 expression in the collecting ducts. Renal vascular hypertension in patients with Alagille syndrome is explained by the widespread vasculopathy and the role of Notch signalling in vascular development. Increased awareness of Alagille syndrome amongst nephrologists may lead to more diagnoses of Alagille syndrome in patients with apparently isolated renal disease.


Assuntos
Síndrome de Alagille/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Nefropatias/metabolismo , Proteínas de Membrana/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais/fisiologia , Síndrome de Alagille/genética , Síndrome de Alagille/fisiopatologia , Proteínas de Ligação ao Cálcio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Nefropatias/genética , Nefropatias/fisiopatologia , Proteínas de Membrana/genética , Receptor Notch2/genética , Proteínas Serrate-Jagged , Transdução de Sinais/genética
17.
J Med Genet ; 50(3): 163-73, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23335808

RESUMO

BACKGROUND: The contribution of copy-number variation (CNV) to disease has been highlighted with the widespread adoption of array-based comparative genomic hybridisation (aCGH) and microarray technology. Contiguous gene deletions involving ANKRD11 in 16q24.3 are associated with autism spectrum disorder (ASD) and intellectual disability (ID), while 16q24.1 deletions affecting FOXF1 are associated with congenital renal malformations, alveolar capillary dysplasia, and various other abnormalities. The disease associations of deletions in the intervening region, 16q24.2, have only been defined to a limited extent. AIM: To determine whether deletions affecting 16q24.2 are correlated with congenital anomalies. METHODS: 35 individuals, each having a deletion in 16q24.2, were characterised clinically and by aCGH and/or SNP-genotyping microarray. RESULTS: Several of the 35 16q24.2 deletions identified here closely abut or overlap the coding regions of FOXF1 and ANKRD11, two genes that have been previously associated with the disease. 25 patients were reported to have ASD/ID, and three were found to have bilateral hydronephrosis. 14 of the deletions associated with ASD/ID overlap the coding regions of FBXO31 and MAP1LC3B. These same genes and two others, C16orf95 and ZCCHC14, are also included in the area of minimal overlap of the three deletions associated with hydronephrosis. CONCLUSIONS: Our data highlight 16q24.2 as a region of interest for ASD, ID and congenital renal malformations. These conditions are associated, albeit without complete penetrance, with deletions affecting C16orf95, ZCCHC14, MAP1LC3B and FBXO31. The function of each gene in development and disease warrants further investigation.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/genética , Cromossomos Humanos Par 16 , Deleção de Genes , Deficiência Intelectual/genética , Rim/anormalidades , Adolescente , Criança , Pré-Escolar , Mapeamento Cromossômico , Variações do Número de Cópias de DNA/genética , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas Repressoras/genética , Adulto Jovem
18.
Pediatr Nephrol ; 28(9): 1757-61, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23052658

RESUMO

Congenital hydronephrosis, defined as congenital dilatation of one or more components of the renal collecting system, is detected in 1-2% of all pregnancies. The majority of antenatally detected hydronephrosis is non-obstructive and either resolves or stabilizes. Management of persistently severe cases may include surgical intervention, the only available treatment. Recent data demonstrate that hedgehog signaling plays a critical role in regulating the structure and function of the collecting system in a manner dependent on the formation of GLI3 repressor. Here, we review the pathobiology and clinical management of non-obstructive hydronephrosis and describe how inhibitors of GLI3 repressor formation may serve as novel therapies for this disorder.


Assuntos
Hidronefrose/congênito , Hidronefrose/terapia , Criança , Proteínas Hedgehog/metabolismo , Humanos , Hidronefrose/genética , Hidronefrose/fisiopatologia , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Sistema Urinário/patologia , Proteína Gli3 com Dedos de Zinco
19.
Nephrology (Carlton) ; 18(3): 161-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23240784

RESUMO

Renal primary cilia are microscopic sensory organelles found on the apical surface of epithelial cells of the nephron and collecting duct. They are based upon a microtubular cytoskeleton, bounded by a specialized membrane, and contain an array of proteins that facilitate their assembly, maintenance and function. Cilium-based signalling is important for the control of epithelial differentiation and has been implicated in the pathogenesis of various cystic kidney diseases and in renal repair. As such, visualizing renal primary cilia and understanding their composition has become an essential component of many studies of inherited kidney disease and mechanisms of epithelial regeneration. Primary cilia were initially identified in the kidney using electron microscopy and this remains a useful technique for the high resolution examination of these organelles. New reagents and techniques now also allow the structure and composition of primary cilia to be analysed in detail using fluorescence microscopy. Primary cilia can be imaged in situ in sections of kidney, and many renal-derived cell lines produce primary cilia in culture providing a simplified and accessible system in which to investigate these organelles. Here we outline microscopy-based techniques commonly used for studying renal primary cilia.


Assuntos
Células Epiteliais/ultraestrutura , Rim/ultraestrutura , Microscopia , Animais , Biomarcadores/análise , Cílios , Células Epiteliais/química , Imunofluorescência , Humanos , Processamento de Imagem Assistida por Computador , Rim/química , Microscopia/métodos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência
20.
J Am Soc Nephrol ; 22(4): 718-31, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21436291

RESUMO

Renal dysplasia, defined by defective ureteric branching morphogenesis and nephrogenesis, is the major cause of renal failure in infants and children. Here, we define a pathogenic role for a ß-catenin-activated genetic pathway in murine renal dysplasia. Stabilization of ß-catenin in the ureteric cell lineage before the onset of kidney development increased ß-catenin levels and caused renal aplasia or severe hypodysplasia. Analysis of gene expression in the dysplastic tissue identified downregulation of genes required for ureteric branching and upregulation of Tgfß2 and Dkk1. Treatment of wild-type kidney explants with TGFß2 or DKK1 generated morphogenetic phenotypes strikingly similar to those observed in mutant kidney tissue. Stabilization of ß-catenin after the onset of kidney development also caused dysplasia and upregulation of Tgfß2 and Dkk1 in the epithelium. Together, these results demonstrate that elevation of ß-catenin levels during kidney development causes dysplasia.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Rim/anormalidades , Rim/embriologia , Fator de Crescimento Transformador beta2/fisiologia , Regulação para Cima/fisiologia , beta Catenina/fisiologia , Animais , Apoptose/fisiologia , Proliferação de Células , Modelos Animais de Doenças , Feminino , Rim/fisiopatologia , Camundongos , Camundongos Mutantes , Morfogênese/fisiologia , Gravidez , Transdução de Sinais/fisiologia , Ureter/anormalidades , Ureter/embriologia , Ureter/fisiopatologia , Proteínas Wnt/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA