Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 326(1): F3-F19, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37916284

RESUMEN

In 1990, mutations of the Wilms' tumor-1 gene (WT1), encoding a transcription factor in the embryonic kidney, were found in 10-15% of Wilms' tumors; germline WT1 mutations were associated with hereditary syndromes involving glomerular and reproductive tract dysplasia. For more than three decades, these discoveries prompted investigators to explore the embryonic role of WT1 and the mechanisms by which loss of WT1 leads to malignant transformation. Here, we discuss how alternative splicing of WT1 generates isoforms that act in a context-specific manner to activate or repress target gene transcription. WT1 also regulates posttranscriptional regulation, alters the epigenetic landscape, and activates miRNA expression. WT1 functions at multiple stages of kidney development, including the transition from resting stem cells to committed nephron progenitor, which it primes to respond to WNT9b signals from the ureteric bud. WT1 then drives nephrogenesis by activating WNT4 expression and directing the development of glomerular podocytes. We review the WT1 mutations that account for Denys-Drash syndrome, Frasier syndrome, and WAGR syndrome. Although the WT1 story began with Wilms' tumors, an understanding of the pathways that link aberrant kidney development to malignant transformation still has some important gaps. Loss of WT1 in nephrogenic rests may leave these premalignant clones with inadequate DNA repair enzymes and may disturb the epigenetic landscape. Yet none of these observations provide a complete picture of Wilms' tumor pathogenesis. It appears that the WT1 odyssey is unfinished and still holds a great deal of untilled ground to be explored.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Humanos , Genes del Tumor de Wilms , Proteínas WT1/genética , Proteínas WT1/metabolismo , Riñón/metabolismo , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Mutación , Neoplasias Renales/genética , Neoplasias Renales/metabolismo
2.
Am J Physiol Renal Physiol ; 324(3): F245-F255, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36546838

RESUMEN

Mammalian nephrons arise from a population of nephron progenitor cells (NPCs) expressing the master transcription factor Wilms tumor-1 (WT1), which is crucial for NPC proliferation, migration, and differentiation. In humans, biallelic loss of WT1 precludes nephrogenesis and leads to the formation of Wilms tumor precursor lesions. We hypothesize that WT1 normally primes the NPC for nephrogenesis by inducing expression of NPC-specific DNA repair genes that protect the genome. We analyzed transcript levels for a panel of DNA repair genes in embryonic day 17.5 (E17.5) versus adult mouse kidneys and noted seven genes that were increased >20-fold. We then isolated Cited1+ NPCs from E17.5 kidneys and found that only one gene, nei-like DNA glycosylase 3 (Neil3), was enriched. RNAscope in situ hybridization of E17.5 mouse kidneys showed increased Neil3 expression in the nephrogenic zone versus mature nephron structures. To determine whether Neil3 expression is WT1 dependent, we knocked down Wt1 in Cited1+ NPCs (60% knockdown efficiency) and noted a 58% reduction in Neil3 transcript levels. We showed that WT1 interacts with the Neil3 promoter and that activity of a Neil3 promoter-reporter vector was increased twofold in WT1+ versus WT1- cells. We propose that Neil3 is a WT1-dependent DNA repair gene expressed at high levels in Cited1+ NPCs, where it repairs mutational injury to the genome during nephrogenesis. NEIL3 is likely just one of many such lineage-specific repair mechanisms that respond to genomic injury during kidney development.NEW & NOTEWORTHY We studied the molecular events leading to Wilms tumors as a model for the repair of genomic injury. Specifically, we showed that WT1 activates DNA repair gene Neil3 in nephron progenitor cells. However, our observations offer a much broader principle, demonstrating that the embryonic kidney invests in lineage-specific expression of DNA repair enzymes. Thus, it is conceivable that failure of these mechanisms could lead to a variety of "sporadic" congenital renal malformations and human disease.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Animales , Humanos , Ratones , Riñón/metabolismo , Neoplasias Renales/patología , Mamíferos/metabolismo , Nefronas/metabolismo , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patología , Proteínas WT1/genética , Proteínas WT1/metabolismo
3.
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
4.
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
5.
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
6.
Pediatr Nephrol ; 34(5): 873-881, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30413946

RESUMEN

BACKGROUND: Cystinosis is an ultrarare disorder caused by mutations of the cystinosin (CTNS) gene, encoding a cystine-selective efflux channel in the lysosomes of all cells of the body. Oral therapy with cysteamine reduces intralysosomal cystine accumulation and slows organ deterioration but cannot reverse renal Fanconi syndrome nor prevent the eventual need for renal transplantation. A definitive therapeutic remains elusive. About 15% of cystinosis patients worldwide carry one or more nonsense mutations that halt translation of the CTNS protein. Aminoglycosides such as geneticin (G418) can bind to the mammalian ribosome, relax translational fidelity, and permit readthrough of premature termination codons to produce full-length protein. METHODS: To ascertain whether aminoglycosides permit readthrough of the most common CTNS nonsense mutation, W138X, we studied the effect of G418 on patient fibroblasts. RESULTS: G418 treatment induced translational readthrough of CTNSW138X constructs transfected into HEK293 cells and expression of full-length endogenous CTNS protein in homozygous W138X fibroblasts. CONCLUSIONS: Reduction in intracellular cystine indicates that the CTNS protein produced is functional as a cystine transporter. Interestingly, similar effects were seen even in W138X compound heterozygotes. These studies establish proof-of-principle for the potential of aminoglycosides to treat cystinosis and possibly other monogenic diseases caused by nonsense mutations.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinosis/tratamiento farmacológico , Fibroblastos/efectos de los fármacos , Gentamicinas/farmacología , Terminación de la Cadena Péptídica Traduccional/efectos de los fármacos , Codón sin Sentido , Cistina/metabolismo , Cistinosis/genética , Fibroblastos/metabolismo , Vectores Genéticos/genética , Gentamicinas/uso terapéutico , Células HEK293 , Humanos , Terminación de la Cadena Péptídica Traduccional/genética , Plásmidos/genética , ARN Mensajero/análisis , Proteínas Recombinantes/genética , Transfección
7.
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
8.
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
9.
Adv Exp Med Biol ; 959: 187-195, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28755196

RESUMEN

In this chapter we describe the current Quebec NTBC Study protocol. Quebec's unique characteristics have influenced the development of the protocol, including a high prevalence of hepatorenal tyrosinemia (HT1), universal newborn screening for HT1, availability of treatment with nitisinone (NTBC) and special diet, a large territory, where HT1 treatment is coordinated by a small number of centers. Screened newborns are seen within 3 weeks of birth. Patients with liver dysfunction (prolonged prothrombin time and/or international normalized ratio (INR) provide sensitive, rapidly available indicators) are treated by NTBC and special diet. The specific diagnosis is confirmed by diagnostic testing for succinylacetone (SA) in plasma and urine samples obtained before treatment. After an initial period of frequent surveillance, stable patients are followed every 3 months by assay of plasma amino acids and NTBC and plasma and urine SA. Abdominal ultrasound is done every 6 months. Patients have an annual visit to the coordinating center that includes multidisciplinary evaluations in metabolic genetics, hepatology, imaging (for abdominal ultrasound and magnetic resonance imaging) and other specialties as necessary. If hepatocellular carcinoma is suspected by imaging and/or because of progressive elevation of alphafetoprotein, liver transplantation is discussed. To date, no patient in whom treatment was started before 1 month of age has developed hepatocellular carcinoma, after surveillance for up to 20 years in some. This patient group is the largest in the world that has been treated rapidly following newborn screening. The protocol continues to evolve to adapt to the challenges of long term surveillance.


Asunto(s)
Ciclohexanonas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Nitrobenzoatos/uso terapéutico , Tirosinemias/tratamiento farmacológico , Heptanoatos/metabolismo , Humanos , Recién Nacido , Hepatopatías/tratamiento farmacológico , Hepatopatías/etiología , Hepatopatías/metabolismo , Trasplante de Hígado/métodos , Tamizaje Neonatal/métodos , Quebec , Tirosinemias/complicaciones , Tirosinemias/metabolismo
10.
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
11.
Am J Physiol Renal Physiol ; 310(10): F1148-56, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26719363

RESUMEN

Focal segmental glomerular sclerosis (FSGS) is an irreversible renal pathology characterized by podocyte detachment from the glomerular basement membrane, hyalinosis, and sclerosis. Clinically, it manifests with proteinuria and progressive loss of glomerular filtration. Primary idiopathic FSGS can occur in isolation and frequently progresses to end-stage renal disease, requiring dialysis or kidney transplantation. In 30-50% of these patients, proteinuria and FSGS recur in the renal allograft, suggesting the presence of a podocyte-toxic factor(s) in the recipient's serum. Currently, there is no reliable way to quantify the serum activity or predict the subset of FSGS patients at risk for recurrence after transplantation. We describe a novel in vitro method that measures the podocyte-toxic activity of sera from FSGS patients using cultured human podocytes; we compare this with the effect of compounds such as adriamycin. Using immunofluorescence microscopy followed by computerized image-processing analysis, we show that incubation of human podocytes with adriamycin leads to a dose-dependent disassembly of focal adhesion complexes (FACs). We then demonstrate that sera from patients with posttransplant recurrent or idiopathic FSGS cause a similar FAC disturbance. In contrast, sera from nonrecurrent FSGS patients do not affect FACs. In some FSGS patients, toxic effects of serum can be prevented by blockade of the tumor necrosis factor-α pathway. We propose that this method may be useful as a diagnostic tool to identify FSGS patients with serum podocyte-toxic activity that presumably places them at increased risk for recurrence in the renal allograft.


Asunto(s)
Adhesiones Focales/efectos de los fármacos , Glomeruloesclerosis Focal y Segmentaria/sangre , Podocitos/efectos de los fármacos , Toxinas Biológicas/sangre , Adolescente , Adulto , Células Cultivadas , Niño , Preescolar , Doxorrubicina , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Reproducibilidad de los Resultados , Medición de Riesgo , Toxinas Biológicas/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
12.
Kidney Int ; 89(6): 1192-203, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27181776

RESUMEN

Nephropathic cystinosis is an autosomal recessive metabolic, lifelong disease characterized by lysosomal cystine accumulation throughout the body that commonly presents in infancy with a renal Fanconi syndrome and, if untreated, leads to end-stage kidney disease (ESKD) in the later childhood years. The molecular basis is due to mutations in CTNS, the gene encoding for the lysosomal cystine-proton cotransporter, cystinosin. During adolescence and adulthood, extrarenal manifestations of cystinosis develop and require multidisciplinary care. Despite substantial improvement in prognosis due to cystine-depleting therapy with cysteamine, no cure of the disease is currently available. Kidney Disease: Improving Global Outcomes (KDIGO) convened a Controversies Conference on cystinosis to review the state-of-the-art knowledge and to address areas of controversies in pathophysiology, diagnostics, monitoring, and treatment in different age groups. More importantly, promising areas of investigation that may lead to optimal outcomes for patients afflicted with this lifelong, systemic disease were discussed with a research agenda proposed for the future.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/genética , Cisteamina/uso terapéutico , Depletores de Cistina/uso terapéutico , Cistina/metabolismo , Cistinosis/etiología , Enfermedades Raras/etiología , Adolescente , Adulto , Factores de Edad , Niño , Congresos como Asunto , Cisteamina/efectos adversos , Depletores de Cistina/efectos adversos , Cistinosis/complicaciones , Cistinosis/diagnóstico , Cistinosis/terapia , Síndrome de Fanconi/complicaciones , Síndrome de Fanconi/tratamiento farmacológico , Pruebas Genéticas , Terapia Genética , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Humanos , Terapia de Inmunosupresión/efectos adversos , Lactante , Fallo Renal Crónico/etiología , Trasplante de Riñón/efectos adversos , Lisosomas/metabolismo , Mutación , Enfermedades Raras/complicaciones , Enfermedades Raras/diagnóstico , Enfermedades Raras/terapia , Diálisis Renal
13.
Nephrol Dial Transplant ; 29 Suppl 4: iv87-94, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25165189

RESUMEN

Cystinosis is caused by mutations in the CTNS gene (17p13.2), which encodes for a lysosomal cystine/proton symporter termed cystinosin. It is the most common cause of inherited renal Fanconi syndrome in young children. Because of its rarity, the diagnosis and specific treatment of cystinosis are frequently delayed, which has a significant impact on the overall prognosis. In this document, we have summarized expert opinions on several aspects of the disease to improve knowledge and provide guidance for diagnosis and treatment.


Asunto(s)
Cistinosis/diagnóstico , Cistinosis/terapia , Niño , Cistinosis/genética , Síndrome de Fanconi/diagnóstico , Síndrome de Fanconi/terapia , Humanos , Guías de Práctica Clínica como Asunto , Sociedades Médicas
14.
Pediatr Nephrol ; 29(4): 705-10, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24414605

RESUMEN

The mammalian kidney arises from OSR1(+) progenitor cells in the intermediate mesoderm. However, these cells must acquire unique properties before they can respond to inductive signals that launch the differentiation program. Recent data indicate that the transcription factor, WT1, plays a master role in this transition. Interestingly, some of these embryonic nephron progenitor cells are retained in the adult organ where they may participate in tissue regeneration after acute kidney injury. A better understanding of the biology of these cells may one day allow progenitor cell-based therapeutic strategies to help regenerate damaged adult nephrons.


Asunto(s)
Diferenciación Celular/fisiología , Riñón/citología , Riñón/embriología , Regeneración/fisiología , Células Madre/citología , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Proteínas WT1/genética , Proteínas WT1/metabolismo
15.
Hum Mol Genet ; 20(21): 4167-74, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21821672

RESUMEN

Human nephrons are formed during fetal life through an interaction between the branching ureteric bud and progenitor cells. The wide variation in final nephron number has been attributed to allelic variants of genes regulating ureteric bud arborization. Here, we hypothesize that dysfunctional variants of the Odd-Skipped Related 1 (OSR1) gene which compromise the renal progenitor cell pool might also limit newborn kidney size and function. We show that OSR1 is expressed in human mesenchymal stem cells, the blastemal component of Wilms tumors and CD24+/CD133+ progenitor cells isolated from the mature kidney. We identified an OSR1(rs12329305(T)) allele in 6% of normal Caucasians which alters an exon2 splice enhancer. This variant is predicted to reduce spliceosome-binding affinity and stability of the OSR1 mRNA. In cultured cells, the OSR1(rs12329305)(T) allele produced no identifiable transcript. Normal Caucasian newborns from Montreal with the OSR1(rs12329305)(T) allele had kidney volume 11.8% smaller (P= 0.006) and cord blood cystatin C levels 12.6% higher (P = 0.005) than those with wild-type genotype. Effects of the OSR1(rs12329305)(T) allele are additive with genes that alter ureteric bud branching. Kidney volume was reduced more in newborns bearing both RET(rs1800860)(A) and OSR1(rs12329305)(T) alleles (22%, P= 0.0008) and cystatin C was increased by 17% (P= 0.006) versus newborns with wild-type alleles. Although only two subjects had PAX2(rs11599825)(A) and OSR1(rs12329305)(T) alleles, kidney size was reduced by 27% and cystatin C was increased by 14% versus wild-types (P= NS).


Asunto(s)
Alelos , Riñón/crecimiento & desarrollo , Riñón/patología , Células Madre Mesenquimatosas/enzimología , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Adulto , Animales , Animales Recién Nacidos , Secuencia de Bases , Separación Celular , Cistatina C/metabolismo , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Humanos , Hibridación in Situ , Recién Nacido , Riñón/enzimología , Ratones , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Tamaño de los Órganos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Cordón Umbilical/metabolismo , Tumor de Wilms/enzimología , Tumor de Wilms/genética , Tumor de Wilms/patología
16.
J Am Soc Nephrol ; 23(1): 63-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22021707

RESUMEN

ß-Catenin/Wnt signaling is essential during early inductive stages of kidney development, but its role during postinductive stages of nephron development and maturation is not well understood. In this study, we used Pax8Cre mice to target ß-catenin deficiency to renal epithelial cells at the late S-shaped body stage and the developing collecting ducts. The conditional ß-catenin knockout mice formed abnormal kidneys and had reduced renal function. The kidneys were hypoplastic with a thin cortex; a superficial layer of tubules was missing. A high proportion of glomeruli had small, underdeveloped capillary tufts. In these glomeruli, well differentiated podocytes replaced parietal epithelial cells in Bowman's capsule; capillaries toward the outer aspect of these podocytes mimicked the formation of glomerular capillaries. Tracing nephrogenesis in embryonic conditional ß-catenin knockout mice revealed that these "parietal podocytes" derived from precursor cells in the parietal layer of the S-shaped body by direct lineage switch. Taken together, these findings demonstrate that ß-catenin/Wnt signaling is important during the late stages of nephrogenesis and for the lineage specification of parietal epithelial cells.


Asunto(s)
Riñón/embriología , Organogénesis , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Epiteliales/citología , Riñón/citología , Riñón/metabolismo , Ratones , Ratones Noqueados , beta Catenina/genética
17.
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
18.
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.

19.
N Engl J Med ; 361(1): 40-51, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19571282

RESUMEN

BACKGROUND: Nephropathy and retinopathy remain important complications of type 1 diabetes. It is unclear whether their progression is slowed by early administration of drugs that block the renin-angiotensin system. METHODS: We conducted a multicenter, controlled trial involving 285 normotensive patients with type 1 diabetes and normoalbuminuria and who were randomly assigned to receive losartan (100 mg daily), enalapril (20 mg daily), or placebo and followed for 5 years. The primary end point was a change in the fraction of glomerular volume occupied by mesangium in kidney-biopsy specimens. The retinopathy end point was a progression on a retinopathy severity scale of two steps or more. Intention-to-treat analysis was performed with the use of linear regression and logistic-regression models. RESULTS: A total of 90% and 82% of patients had complete renal-biopsy and retinopathy data, respectively. Change in mesangial fractional volume per glomerulus over the 5-year period did not differ significantly between the placebo group (0.016 units) and the enalapril group (0.005, P=0.38) or the losartan group (0.026, P=0.26), nor were there significant treatment benefits for other biopsy-assessed renal structural variables. The 5-year cumulative incidence of microalbuminuria was 6% in the placebo group; the incidence was higher with losartan (17%, P=0.01 by the log-rank test) but not with enalapril (4%, P=0.96 by the log-rank test). As compared with placebo, the odds of retinopathy progression by two steps or more was reduced by 65% with enalapril (odds ratio, 0.35; 95% confidence interval [CI], 0.14 to 0.85) and by 70% with losartan (odds ratio, 0.30; 95% CI, 0.12 to 0.73), independently of changes in blood pressure. There were three biopsy-related serious adverse events that completely resolved. Chronic cough occurred in 12 patients receiving enalapril, 6 receiving losartan, and 4 receiving placebo. CONCLUSIONS: Early blockade of the renin-angiotensin system in patients with type 1 diabetes did not slow nephropathy progression but slowed the progression of retinopathy. (ClinicalTrials.gov number, NCT00143949.)


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Enalapril/farmacología , Glomérulos Renales/efectos de los fármacos , Losartán/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Retina/efectos de los fármacos , Adulto , Albuminuria , Bloqueadores del Receptor Tipo 1 de Angiotensina II/efectos adversos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Nefropatías Diabéticas/prevención & control , Retinopatía Diabética/prevención & control , Progresión de la Enfermedad , Método Doble Ciego , Enalapril/efectos adversos , Enalapril/uso terapéutico , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Estimación de Kaplan-Meier , Glomérulos Renales/patología , Modelos Logísticos , Losartán/efectos adversos , Losartán/uso terapéutico , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/patología , Retina/patología
20.
Mol Genet Metab ; 107(1-2): 49-54, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22885033

RESUMEN

BACKGROUND: Hepatorenal tyrosinemia (HT1, fumarylacetoacetate hydrolase deficiency, MIM 276700) can cause severe hepatic, renal and peripheral nerve damage. In Québec, HT1 is frequent and neonatal HT1 screening is practiced. Nitisinone (NTBC, Orfadin ®) inhibits tyrosine degradation prior to the formation of toxic metabolites like succinylacetone and has been offered to HT1 patients in Québec since 1994. METHODS: We recorded the clinical course of 78 Québec HT1 patients born between 1984 and 2004. There were three groups: those who never received nitisinone (28 patients), those who were first treated after 1 month of age (26 patients) and those treated before 1 month (24 patients). Retrospective chart review was performed for events before 1994, when nitisinone treatment began, and prospective data collection thereafter. FINDINGS: No hospitalizations for acute complications of HT1 occurred during 5731 months of nitisinone treatment, versus 184 during 1312 months without treatment (p<0.001). Liver transplantation was performed in 20 non-nitisinone-treated patients (71%) at a median age of 26 months, versus 7 late-treated patients (26%, p<0.001), and no early-treated patient (p<0.001). No early-treated patient has developed detectable liver disease after more than 5 years. Ten deaths occurred in non-nitisinone treated patients versus two in treated patients (p<0.01). Both of the latter deaths were from complications of transplantation unrelated to HT1. One probable nitisinone-related event occurred, transient corneal crystals with photophobia. INTERPRETATION: Nitisinone treatment abolishes the acute complications of HT1. Some patients with established liver disease before nitisinone treatment eventually require hepatic transplantation. Patients who receive nitisinone treatment before 1 month had no detectable liver disease after more than 5 years.


Asunto(s)
Ciclohexanonas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Nitrobenzoatos/uso terapéutico , Tirosinemias/tratamiento farmacológico , Niño , Preescolar , Ciclohexanonas/efectos adversos , Inhibidores Enzimáticos/efectos adversos , Humanos , Lactante , Recién Nacido , Riñón/metabolismo , Hígado/metabolismo , Trasplante de Hígado , Tamizaje Neonatal , Nitrobenzoatos/efectos adversos , Quebec , Resultado del Tratamiento , Tirosinemias/diagnóstico , Tirosinemias/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA