Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Curr Opin Nephrol Hypertens ; 32(3): 297-304, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37013453

RESUMEN

PURPOSE OF REVIEW: Servais et al. recently published clinical practice recommendations for the care of cystinuria patients. However, these guidelines were largely based on retrospective data from adults and children presenting with stones. Significant questions remain about the natural history of cystinuria in presymptomatic children. RECENT FINDINGS: We review the natural history of cystinuria in presymptomatic children followed from birth. In total, 130 pediatric patients were assigned putative genotypes based on parental urinary phenotype: type A/A (N = 23), B/B (N = 6), and B/N (N = 101). Stones were identified in 12/130 (4% of A/A, 17% of B/B, and 1% of B/N patients). Type B/B patients had lower cystine excretion than type A/A patients. Although urine cystine/creatinine fell with age, urine cystine/l rose progressively in parallel with the risk of nephrolithiasis. Each new stone was preceded by 6-12 months of urine specific gravity of more than 1.020. However, average urine specific gravity and pH were not different in stone formers vs. nonstone formers, suggesting that intrinsic stone inhibitors or other unknown factors may be the strongest determinants of individual risk. SUMMARY: The current study reviews the clinical evolution of cystinuria in a cohort of children identified by newborn screening, who were categorized by urinary phenotype and followed from birth.


Asunto(s)
Cistinuria , Cálculos Renales , Humanos , Cistinuria/diagnóstico , Cistinuria/genética , Cistinuria/orina , Estudios Retrospectivos , Cistina/genética , Cálculos Renales/orina , Fenotipo
2.
Pediatr Nephrol ; 35(11): 2031-2042, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31807928

RESUMEN

The advent of a new class of aminoglycosides with increased translational readthrough of nonsense mutations and reduced toxicity offers a new therapeutic strategy for a subset of patients with hereditary kidney disease. The renal uptake and retention of aminoglycosides at a high intracellular concentration makes the kidney an ideal target for this approach. In this review, we explore the potential of aminoglycoside readthrough therapy in a number of hereditary kidney diseases and discuss the therapeutic window of opportunity for subclasses of each disease, when caused by nonsense mutations.


Asunto(s)
Aminoglicósidos/uso terapéutico , Antibacterianos/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Aminoglicósidos/farmacocinética , Antibacterianos/farmacocinética , Codón sin Sentido/efectos de los fármacos , Humanos , Enfermedades Renales/genética
3.
Kidney Int ; 96(4): 850-861, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31420194

RESUMEN

The podocyte is a key component of the glomerular filtration barrier. Podocyte dysfunction is central to the underlying pathophysiology of many common glomerular diseases, including diabetic nephropathy, glomerulonephritis and genetic forms of nephrotic syndrome. Collectively, these conditions affect millions of people worldwide, and account for the majority of kidney diseases requiring dialysis and transplantation. The 12th International Podocyte Conference was held in Montreal, Canada from May 30 to June 2, 2018. The primary aim of this conference was to bring together nephrologists, clinician scientists, basic scientists and their trainees from all over the world to present their research and to establish networks with the common goal of developing new therapies for glomerular diseases based on the latest advances in podocyte biology. This review briefly highlights recent advances made in understanding podocyte structure and metabolism, experimental systems in which to study podocytes and glomerular disease, disease mediators, genetic and immune origins of glomerulopathies, and the development of novel therapeutic agents to protect podocyte and glomerular injury.


Asunto(s)
Nefropatías Diabéticas/terapia , Barrera de Filtración Glomerular/fisiopatología , Glomerulonefritis/terapia , Síndrome Nefrótico/terapia , Podocitos/patología , Animales , Canadá , Congresos como Asunto , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Glomerulonefritis/patología , Glomerulonefritis/fisiopatología , Humanos , Síndrome Nefrótico/patología , Síndrome Nefrótico/fisiopatología
4.
Pediatr Nephrol ; 33(10): 1741-1750, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29982877

RESUMEN

BACKGROUND: Children with genetic causes of steroid-resistant nephrotic syndrome (SRNS) usually do well after renal transplantation, while some with idiopathic SRNS show recurrence due to a putative podocyte-toxic factor. Distinguishing different forms of SRNS based on clinical criteria has been difficult. The aim of our study was to test a novel approach that allows categorization of patients into clinically useful subgroups. METHODS: Seventeen patients with clinically confirmed SRNS were analyzed by next-generation sequencing (NGS) of 37 known SRNS genes and a functional assay of cultured human podocytes, which indirectly tests for toxicity of patients' sera by evidenced loss of podocyte focal adhesion complex (FAC) number. RESULTS: We identified a pathogenic mutation in seven patients (41%). Sera from patients with monogenic SRNS caused mild loss of FAC number down to 73% compared to untreated controls, while sera from seven of the remaining ten patients with idiopathic SRNS caused significant FAC number loss to 43% (non-overlapping difference 30%, 95% CI 26-36%, P < 0.001). All patients with recurrent SRNS (n = 4) in the graft showed absence of podocyte gene mutations but significant FAC loss. Three patients had no mutation nor serum podocyte toxicity. CONCLUSIONS: Our approach allowed categorization of patients into three subgroups: (1) patients with monogenic SRNS; (2) patients with idiopathic SRNS and marked serum podocyte toxicity; and (3) patients without identifiable genetic cause nor evidence of serum podocyte toxicity. Post-transplant SRNS recurrence risk appears to be low in groups 1 and 3, but high in group 2.


Asunto(s)
Resistencia a Medicamentos/genética , Glucocorticoides/farmacología , Síndrome Nefrótico/genética , Podocitos/patología , Adolescente , Adulto , Técnicas de Cultivo de Célula , Línea Celular , Niño , Preescolar , Femenino , Glucocorticoides/uso terapéutico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología , Recurrencia , Estudios Retrospectivos , Adulto Joven
5.
J Biol Chem ; 291(8): 3785-95, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26655220

RESUMEN

Hereditary forms of Wilms arise from developmentally arrested clones of renal progenitor cells with biallelic mutations of WT1; recently, it has been found that Wilms tumors may also be associated with biallelic mutations in DICER1 or DROSHA, crucial for miRNA biogenesis. We have previously shown that a critical role for WT1 during normal nephrogenesis is to suppress transcription of the Polycomb group protein, EZH2, thereby de-repressing genes in the differentiation cascade. Here we show that WT1 also suppresses translation of EZH2. All major WT1 isoforms induce an array of miRNAs, which target the 3' UTR of EZH2 and other Polycomb-associated transcripts. We show that the WT1(+KTS) isoform binds to the 5' UTR of EZH2 and interacts directly with the miRNA-containing RISC to enhance post-transcriptional inhibition. These observations suggest a novel mechanism through which WT1 regulates the transition from resting stem cell to activated progenitor cell during nephrogenesis. Our findings also offer a plausible explanation for the fact that Wilms tumors can arise either from loss of WT1 or loss of miRNA processing enzymes.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Complejo Represivo Polycomb 2/biosíntesis , Biosíntesis de Proteínas , ARN Neoplásico/metabolismo , Proteínas WT1/metabolismo , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Células Madre Mesenquimatosas/patología , MicroARNs/genética , Complejo Represivo Polycomb 2/genética , ARN Neoplásico/genética , Proteínas WT1/genética , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patología
6.
J Biol Chem ; 290(4): 2279-88, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25331950

RESUMEN

The mammalian kidney is derived from progenitor cells in intermediate mesoderm. During embryogenesis, progenitor cells expressing the Wilms tumor suppressor gene, WT1, are induced to differentiate in response to WNT signals from the ureteric bud. In hereditary Wilms tumors, clonal loss of WT1 precludes the ß-catenin pathway response and leads to precancerous nephrogenic rests. We hypothesized that WT1 normally primes progenitor cells for differentiation by suppressing the enhancer of zeste2 gene (EZH2), involved in epigenetic silencing of differentiation genes. In human amniotic fluid-derived mesenchymal stem cells, we show that exogenous WT1B represses EZH2 transcription. This leads to a dramatic decrease in the repressive lysine 27 trimethylation mark on histone H3 that silences ß-catenin gene expression. As a result, amniotic fluid mesenchymal stem cells acquire responsiveness to WNT9b and increase expression of genes that mark the onset of nephron differentiation. Our observations suggest that biallelic loss of WT1 sustains the inhibitory histone methylation state that characterizes Wilms tumors.


Asunto(s)
Amnios/metabolismo , Epigénesis Genética , Histonas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteínas WT1/metabolismo , beta Catenina/genética , Secuencias de Aminoácidos , Células Cultivadas , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Humanos , Células Madre Mesenquimatosas/citología , Embarazo , Células Madre/citología , Tumor de Wilms/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
7.
Eur J Hum Genet ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987663

RESUMEN

Biallelic pathogenic variants in the gene CC2D2A cause a spectrum of ciliopathies, including Joubert and Meckel syndrome, which frequently involve the kidney; however, no cases of isolated renal disease (i.e., nephronophthisis) have yet been reported. In an adult with a clinical presentation consistent with nephronophthisis, next-generation sequencing identified a rare homozygous nonsense variant in CC2D2A (c.100 C > T; p.(Arg34*)). Tissue-specific expression data and promoter activity analysis demonstrates that this variant primarily affects a transcript isoform predominant in the kidneys but does not affect the transcript isoforms predominant in other tissues typically involved in CC2D2A-related ciliopathies (e.g., cerebellum, liver). Expression analysis of patient-specific cDNA in MDCK cells demonstrates partial translation re-initiation downstream of p.(Arg34*) as a possible escape mechanism from nonsense mediated decay. These data provide mechanistic insights in support of this novel genotype-phenotype association.

8.
Semin Nephrol ; 43(4): 151437, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37968178

RESUMEN

The tubular system of the kidneys is a complex series of morphologic and functional units orchestrating the content of tubular fluid as it flows along the nephron and collecting ducts. Renal tubules maintain body water, regulate electrolytes and acid-base balance, reabsorb precious organic solutes, and eliminate specific metabolites, toxins, and drugs. In addition, decisive mechanisms to adjust blood pressure are governed by the renal tubules. Genetic as well as acquired disorders of these tubular functions may cause serious diseases that manifest both in childhood and adulthood. This article addresses a selection of tubulopathies and the underlying pathomechanisms, while highlighting the important differences in pediatric and adult nephrology care. These range from rare monogenic conditions such as nephrogenic diabetes insipidus, cystinosis, and Bartter syndrome that present in childhood, to the genetic and acquired tubular pathologies causing hypertension or nephrolithiasis that are more prevalent in adults. Both pediatric and adult nephrologists must be aware of these conditions and the age-dependent manifestations that warrant close interaction between the two subspecialties.


Asunto(s)
Diabetes Insípida Nefrogénica , Nefrología , Humanos , Niño , Túbulos Renales/patología , Riñón/patología , Diabetes Insípida Nefrogénica/genética , Diabetes Insípida Nefrogénica/patología , Nefronas
9.
Can J Kidney Health Dis ; 10: 20543581231191836, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37564323

RESUMEN

Purpose of program: Adolescents and young adults with chronic disease face many personal and systemic barriers that may impede their successful transition from pediatric to adult care, putting them at risk for treatment nonadherence, loss to follow-up, and poor health outcomes. Such barriers include impaired socioemotional functioning, overreliance on adult caregivers, lack of disease-specific knowledge, and poor coordination between pediatric and adult health care services. In 2007, we established a specialized youth to adult nephrology transition clinic at a tertiary care center to address these barriers and provide adolescents and young adults with renal disease followed at the affiliated children's hospital with a seamless transition to adult care. Sources of information: The attending clinic nephrologist collected data prospectively for this quality improvement report. Methods: The features of this specialized clinic included (1) single point of entry and single triage adult nephrologist, (2) ongoing follow-up with a single adult nephrologist who communicated with the pediatric nephrologists, and (3) a single specialized clinic nurse who provided disease-specific education and helped to ensure ongoing patient engagement and follow-up. Importantly, the transition patients were booked into regular appointment slots in the adult nephrologist's general clinic, which facilitated regular follow-up without additional resources. The salary of the transition clinic nurse was covered by an unrestricted grant. Patient visits were in-person, except between 2020 and 2021 when visits were by telephone due to the pandemic. Key findings: A total of 213 patients were referred and assessed in the transition clinic from February 2007 until October 2022. Most referrals were from pediatric nephrologists. Among the patients, 29% had a hereditary kidney disease; in 71%, the disease was acquired. The most common disease was glomerulonephritis and ~30% of the patients suffered from a "rare" disease. Of the 213 patients, 123 (58%) continue to be followed up (mean follow-up: 4.8 years), 27 (13%) were transferred to other physicians, in part to accommodate treatment closer to patients' homes, and 29 (14%) without ongoing care needs were discharged. Only 33 (15%) were lost to follow-up. There were several advantages to the clinic, including the maintenance of accurate records, a process to minimize loss to follow-up, and a "critical mass" of patients with rare diseases, which facilitated development of special expertise in rare disease pathogenesis, diagnosis, treatment, and management of complications. Patients with glomerulonephritis demonstrated a stable serum creatinine over 3 to 15 years, and morbidity (as reflected by emergency room visits and hospitalizations) was low. Limitations: Due to the relatively small numbers of patients in the disease categories, it was not possible to determine conclusively whether attendance of patients in the transition clinic reduced the rate of progression of kidney disease or morbidity. Implications: A dedicated referral, triage, and follow-up process post-transition with only modest financial resources and personnel can result in accurate tracking of clinic data, as well as consistent and reliable follow-up and expert patient care.


Objectif du programme: Les adolescents et les jeunes adultes souffrant de maladies chroniques sont confrontés à de nombreux obstacles, tant personnels que systémiques, qui peuvent entraver leur transition des soins pédiatriques vers les soins aux adultes, ce qui augmente le risque d'inobservance du traitement, de perte de suivi et de mauvais résultats de santé. Parmi ces obstacles, on compte notamment un fonctionnement socioémotionnel déficient, une dépendance excessive envers leurs soignants adultes, le manque de connaissances spécifiques sur leur maladie et une mauvaise coordination entre les services de soins de santé pédiatriques et pour adultes. En 2007, dans un centre de soins tertiaires, nous avons créé une clinique spécialisée dans la transition entre les soins de néphrologie pédiatriques et les soins pour adultes; ceci afin d'éliminer les obstacles et de fournir aux adolescents et aux jeunes adultes qui étaient suivis à l'hôpital pour enfants affilié de faire une transition sans heurt vers les soins aux adultes. Sources: Le néphrologue de la clinique a recueilli des données de façon prospective pour la rédaction de ce rapport d'amélioration de la qualité. Méthodologie: La clinique spécialisée comportait les particularités suivantes: i) un seul point d'entrée et un seul néphrologue pour adultes procédait au triage, ii) un suivi continu était effectué par un seul néphrologue pour adultes qui avait communiqué avec les néphrologues pédiatriques, et iii) une seule infirmière clinicienne spécialisée fournissait de l'information spécifique à la maladie et contribuait à assurer la constance dans l'engagement des patients et leur suivi. Surtout, les patients en transition avaient été inscrits pour des rendez-vous réguliers à la clinique générale du néphrologue pour adultes, ce qui a facilité un suivi sans besoin de ressources supplémentaires. Le salaire de l'infirmière de la clinique de transition était couvert par une subvention illimitée. Les visites des patients se faisaient en personne, sauf en 2020-2021, où les rencontres se faisaient par téléphone en raison de la pandémie. Principaux résultats: Au total, 213 patients ont été aiguillés et évalués à la clinique de transition entre février 2007 et octobre 2022. La plupart des aiguillages provenaient de néphrologues pédiatriques. Parmi les patients, 29 % présentaient une néphropathie héréditaire; 71 % avaient une maladie acquise. La glomérulonéphrite était la néphropathie la plus fréquente, et environ 30 % des patients souffraient d'une maladie dite « rare ¼. Sur les 213 patients aiguillés vers la clinique, 123 (58 %) continuent d'y être suivis (suivi moyen de 4,8 ans), 27 (13 %) ont été transférés à d'autres médecins, en partie pour recevoir des soins plus près de leur domicile, et 29 (14 %) sans besoins de soins continus ont reçu leur congé. Seuls 33 patients (15 %) ont été perdus en cours de suivi. La clinique a présenté plusieurs avantages: une tenue précise des dossiers, un processus visant à minimiser la perte de suivi et une « masse critique ¼ de patients atteints de maladies rares, ce qui a facilité le développement d'une expertise particulière dans leur pathogenèse, leur diagnostic, leur traitement et la prise en charge des leurs complications. Les patients atteints de glomérulonéphrite ont conservé un taux de créatinine sérique stable sur 3 à 15 ans, et la morbidité (reflétée par les visites aux urgences et les hospitalisations) était faible. Limites: Le nombre relativement faible de patients dans les différentes catégories de maladies n'a pas permis de déterminer de façon concluante si le suivi des patients à la clinique de transition avait réduit le taux de progression de la maladie ou la morbidité. Conclusion: La présence d'un processus d'aiguillage, de triage et de suivi dédié après la transition, même en disposant de personnel et de ressources financières modestes, peut se traduire par un suivi précis des données cliniques, ainsi que par un suivi cohérent et fiable et des soins spécialisés pour les patients.

10.
Kidney Int ; 80(2): 146-53, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21389971

RESUMEN

During murine kidney development, canonical WNT signaling is highly active in tubules until about embryonic days E16-E18. At this time, ß-catenin transcriptional activity is progressively restricted to the nephrogenic zone. The cilial protein genes PKD1 and PKD2 are known to be mutated in autosomal dominant polycystic kidney disease (ADPKD), and previous studies proposed that these mutations could lead to a failure to suppress canonical WNT signaling activity. Several in vitro studies have found a link between cilial signaling and ß-catenin regulation, suggesting that aberrant activity might contribute to the cystic phenotype. To study this, we crossed T-cell factor (TCF)/ß-catenin-lacZ reporter mice with mice having Pkd1 or Pkd2 mutations and found that there was no ß-galactosidase staining in cells lining the renal cysts. Thus, suppression of canonical WNT activity, defined by the TCF/ß-catenin-lacZ reporter, is normal in these two different models of polycystic kidney disease. Hence, excessive ß-catenin transcriptional activity may not contribute to cystogenesis in these models of ADPKD.


Asunto(s)
Quistes/etiología , Riñón Poliquístico Autosómico Dominante/metabolismo , Factores de Transcripción TCF/genética , beta Catenina/metabolismo , Factores de Edad , Animales , Túbulos Renales/metabolismo , Ratones , Ratones Mutantes , Modelos Biológicos , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Factores de Transcripción TCF/metabolismo , Canales Catiónicos TRPP/genética , Transcripción Genética , Proteínas Wnt/metabolismo
11.
Pediatr Clin North Am ; 66(1): 179-207, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30454743

RESUMEN

Hypophosphatemic rickets, mostly of the X-linked dominant form caused by pathogenic variants of the PHEX gene, poses therapeutic challenges with consequences for growth and bone development and portends a high risk of fractions and poor bone healing, dental problems and nephrolithiasis/nephrocalcinosis. Conventional treatment consists of PO4 supplements and calcitriol requiring monitoring for treatment-emergent adverse effects. FGF23 measurement, where available, has implications for the differential diagnosis of hypophosphatemia syndromes and, potentially, treatment monitoring. Newer therapeutic modalities include calcium sensing receptor modulation (cinacalcet) and biological molecules targeting FGF23 or its receptors. Their long-term effects must be compared with those of conventional treatments.


Asunto(s)
Raquitismo Hipofosfatémico/diagnóstico , Raquitismo Hipofosfatémico/tratamiento farmacológico , Raquitismo Hipofosfatémico/genética , Calcimiméticos/uso terapéutico , Niño , Diagnóstico Diferencial , Factor-23 de Crecimiento de Fibroblastos , Hormona del Crecimiento/uso terapéutico , Humanos , Mutación , Fosfatos/uso terapéutico , Vitamina D/uso terapéutico
12.
Pediatr Dev Pathol ; 12(5): 347-54, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18666806

RESUMEN

Wilms tumor (WT) is the most frequent renal neoplasm of childhood; a myogenic component is observed in 5% to 10% of tumors. We demonstrate for the first time that myogenic WTs are associated with expression of PAX3, a transcription factor known to specify myoblast cell fate during muscle development. In a panel of 20 WTs, PAX3 was identified in 13 of 13 tumor samples with myogenic histopathology but was absent in 7 of 7 tumors lacking a myogenic component. Furthermore, we show that PAX3 is expressed in the metanephric mesenchyme and stromal compartment of developing mouse kidney. Modulation of endogenous PAX3 expression in human embryonic kidney (HEK293) cells influenced cell migration in in vitro assays. Mutations of WT1 were consistently associated with PAX3 expression in WTs, and modulation of WT1 expression in HEK293 cells was inversely correlated with the level of endogenous PAX3 protein. We demonstrate abundant PAX3 and absence of PAX2 expression in a novel cell line (WitP3) isolated from the stromal portion of a WT bearing a homozygous deletion of the WT1 gene. We hypothesize that PAX3 sets stromal cell fate in developing kidney but is normally suppressed by WT1 during the mesenchyme-to-epithelium transition leading to nephrogenesis. Loss of WT1 permits aberrant PAX3 expression in a subset of WTs with myogenic phenotype.


Asunto(s)
Neoplasias Renales/metabolismo , Riñón/metabolismo , Factores de Transcripción Paired Box/biosíntesis , Tumor de Wilms/metabolismo , Animales , Western Blotting , Diferenciación Celular , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Riñón/embriología , Neoplasias Renales/genética , Neoplasias Renales/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mesodermo/metabolismo , Ratones , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Tumor de Wilms/genética , Tumor de Wilms/patología
13.
Am J Physiol Renal Physiol ; 293(2): F494-500, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17494089

RESUMEN

The canonical WNT signaling pathway plays a crucial role in patterning of the embryo during development, but little is known about the specific developmental events which are under WNT control. To understand more about how the WNT pathway orchestrates mammalian organogenesis, we studied the canonical beta-catenin-mediated WNT signaling pathway in kidneys of mice bearing a beta-catenin-responsive TCF/betaGal reporter transgene. In metanephric kidney, intense canonical WNT signaling was evident in epithelia of the branching ureteric bud and in nephrogenic mesenchyme during its transition into renal tubules. WNT signaling activity is rapidly downregulated in maturing nephrons and becomes undetectable in postnatal kidney. Sites of TCF/betaGal activity are in proximity to the known sites of renal WNT2b and WNT4 expression, and these WNTs stimulate TCF reporter activity in kidney cell lines derived from ureteric bud and metanephric mesenchyme lineages. When fetal kidney explants from HoxB7/GFP mice were exposed to the canonical WNT signaling pathway inhibitor, Dickkopf-1, arborization of the ureteric bud was significantly reduced. We conclude that restricted zones of intense canonical WNT signaling drive branching nephrogenesis in fetal kidney.


Asunto(s)
Riñón/embriología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Animales , Línea Celular , ADN Complementario/biosíntesis , ADN Complementario/genética , Regulación hacia Abajo/fisiología , Células Epiteliales/fisiología , Genes Reporteros/genética , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/metabolismo , Túbulos Renales Colectores/metabolismo , Operón Lac/genética , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Factores del Dominio POU/genética , Transfección , Uréter/embriología , beta Catenina/metabolismo
14.
Hum Mol Genet ; 15(24): 3520-8, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17082250

RESUMEN

Mutations in PKD1 cause dominant polycystic kidney disease (PKD), characterized by large fluid-filled kidney cysts in adult life, but the molecular mechanism of cystogenesis remains obscure. Ostrom et al. [Dev. Biol., 219, 250-258 (2000)] showed that reduced dosage of Pax2 caused increased apoptosis, and ameliorated cystogenesis in Cpk mutant mice with recessive PKD. Pax2 is expressed in condensing metanephrogenic mesenchyme and arborizing ureteric bud, and plays an important role in kidney development. Transient Pax2 expression during fetal kidney mesenchyme-to-epithelial transition, as well as in nascent tubules, is followed by marked down-regulation of Pax2 expression. Here, we show that in humans with PKD, as well as in Pkd1(del34/del34) mutant mice, Pax2 was expressed in cyst epithelial cells, and facilitated cyst growth in Pkd1(del34/del34) mutant mice. In Pkd1(del34/del34) mutant kidneys, the expression of Pax2 persisted in nascent collecting ducts. In contrast, homozygous Pkd1(del34/del34) fetal mice carrying mutant Pax2 exhibited ameliorated cyst growth, although reduced cystogenesis was not associated with increased apoptosis. Pax2 expression was attenuated in nascent collecting ducts and absent from remnant cysts of Pkd1(del34/del34)/Pax2(1Neu/+) mutant mice. To investigate whether the Pkd1 gene product, Polycystin-1, regulates Pax2, MDCK cells were engineered constitutively expressing wild-type Pkd1; Pax2 protein levels and promoter activity were both repressed in MDCK cells over-expressing Pkd1, but not in cells without transgenic Pkd1. These data suggest that polycystin-1-deficient tubular epithelia persistently express Pax2 in ADPKD, and that Pax2 or its pathway may be an appropriate target for the development of novel therapies for ADPKD.


Asunto(s)
Dosificación de Gen , Factor de Transcripción PAX2/genética , Riñón Poliquístico Autosómico Dominante/genética , Animales , Apoptosis , Western Blotting , Línea Celular , Proliferación Celular , Heterocigoto , Homocigoto , Humanos , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Mutantes , Factor de Transcripción PAX2/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Pirofosfatasas/genética , Pirofosfatasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA