Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Nephrol Ther ; 16(7): 420-423, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33162364

RESUMEN

AIM: Aim of the study was to determine if carotid intima media thickness in children with idiopathic nephrotic syndrome is greater than in healthy subjects, and to assess whether carotid intima media thickness in children with nephrotic syndrome is associated with clinical (including disease duration, cumulative dose of steroids, number of relapses) and biochemical parameters. METHODS: A cross-sectional study included 40 patients with nephrotic syndrome (mean age 11.7±4.7 years). Steroid dependent nephrotic syndrome was established in 32 patients (80%), while 8 (20%) had steroid resistant nephrotic syndrome. Control group consisted of 20 age and gender matched healthy children. Blood pressure based on 24-h ambulatory blood pressure monitoring (ABPM), carotid intima media thickness, fasting glucose, insulin, HbA1c, lipid concentrations were measured in all children. RESULTS: A significant difference was detected in carotid intima media thickness values (P=0.036). Children with nephrotic syndrome had significantly greater carotid intima media thickness compared with healthy children (0.42±0.06 and 0.38±0.03mm). Carotid intima-media thickness was positively associated with duration of nephrotic syndrome (r=0.45; P=0.004), body mass index (r=0.48; P=0.002), daytime systolic blood pressure (r=0.46; P=0.003) and night-time systolic blood pressure (r=0.52; P=0.001). Multiple linear regression showed that duration of nephrotic syndrome was the only independent predictor of carotid intima media thickness in children with nephrotic syndrome (R2=0.244; ß=0.327; P=0.037). CONCLUSION: The findings of the present study suggest subclinical vascular damage in patients with nephrotic syndrome. Duration of nephrotic syndrome was the only independent predictor of carotid intima media thickness.


Asunto(s)
Grosor Intima-Media Carotídeo , Síndrome Nefrótico/complicaciones , Presión Sanguínea , Índice de Masa Corporal , Estudios de Casos y Controles , Niño , Estudios Transversales , Femenino , Humanos , Masculino , Sístole
2.
Hum Genet ; 138(10): 1105-1115, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31230195

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease (~ 45%) that manifests before 30 years of age. The genetic locus containing COL4A1 (13q33-34) has been implicated in vesicoureteral reflux (VUR), but mutations in COL4A1 have not been reported in CAKUT. We hypothesized that COL4A1 mutations cause CAKUT in humans. We performed whole exome sequencing (WES) in 550 families with CAKUT. As negative control cohorts we used WES sequencing data from patients with nephronophthisis (NPHP) with no genetic cause identified (n = 257) and with nephrotic syndrome (NS) due to monogenic causes (n = 100). We identified a not previously reported heterozygous missense variant in COL4A1 in three siblings with isolated VUR. When examining 549 families with CAKUT, we identified nine additional different heterozygous missense mutations in COL4A1 in 11 individuals from 11 unrelated families with CAKUT, while no COL4A1 mutations were identified in a control cohort with NPHP and only one in the cohort with NS. Most individuals (12/14) had isolated CAKUT with no extrarenal features. The predominant phenotype was VUR (9/14). There were no clinical features of the COL4A1-related disorders (e.g., HANAC syndrome, porencephaly, tortuosity of retinal arteries). Whereas COL4A1-related disorders are typically caused by glycine substitutions in the collagenous domain (84.4% of variants), only one variant in our cohort is a glycine substitution within the collagenous domain (1/10). We identified heterozygous COL4A1 mutations as a potential novel autosomal dominant cause of CAKUT that is allelic to the established COL4A1-related disorders and predominantly caused by non-glycine substitutions.


Asunto(s)
Colágeno Tipo IV/genética , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/genética , Riñón/anomalías , Mutación , Fenotipo , Sistema Urinario/anomalías , Alelos , Sustitución de Aminoácidos , Biología Computacional/métodos , Análisis Mutacional de ADN , Bases de Datos Genéticas , Evolución Molecular , Femenino , Estudios de Asociación Genética , Sitios Genéticos , Genómica/métodos , Heterocigoto , Humanos , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/genética , Masculino , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Navegador Web , Secuenciación del Exoma
3.
J Am Soc Nephrol ; 29(9): 2348-2361, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30143558

RESUMEN

BACKGROUND: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most prevalent cause of kidney disease in the first three decades of life. Previous gene panel studies showed monogenic causation in up to 12% of patients with CAKUT. METHODS: We applied whole-exome sequencing to analyze the genotypes of individuals from 232 families with CAKUT, evaluating for mutations in single genes known to cause human CAKUT and genes known to cause CAKUT in mice. In consanguineous or multiplex families, we additionally performed a search for novel monogenic causes of CAKUT. RESULTS: In 29 families (13%), we detected a causative mutation in a known gene for isolated or syndromic CAKUT that sufficiently explained the patient's CAKUT phenotype. In three families (1%), we detected a mutation in a gene reported to cause a phenocopy of CAKUT. In 15 of 155 families with isolated CAKUT, we detected deleterious mutations in syndromic CAKUT genes. Our additional search for novel monogenic causes of CAKUT in consanguineous and multiplex families revealed a potential single, novel monogenic CAKUT gene in 19 of 232 families (8%). CONCLUSIONS: We identified monogenic mutations in a known human CAKUT gene or CAKUT phenocopy gene as the cause of disease in 14% of the CAKUT families in this study. Whole-exome sequencing provides an etiologic diagnosis in a high fraction of patients with CAKUT and will provide a new basis for the mechanistic understanding of CAKUT.


Asunto(s)
Secuenciación del Exoma/métodos , Predisposición Genética a la Enfermedad/epidemiología , Linaje , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Animales , Humanos , Incidencia , Riñón/anomalías , Ratones , Fenotipo , Pronóstico , Medición de Riesgo , Sensibilidad y Especificidad , Distribución por Sexo , Sistema Urinario/anomalías , Anomalías Urogenitales/epidemiología , Reflujo Vesicoureteral/epidemiología
4.
Nat Commun ; 9(1): 1960, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773874

RESUMEN

No efficient treatment exists for nephrotic syndrome (NS), a frequent cause of chronic kidney disease. Here we show mutations in six different genes (MAGI2, TNS2, DLC1, CDK20, ITSN1, ITSN2) as causing NS in 17 families with partially treatment-sensitive NS (pTSNS). These proteins interact and we delineate their roles in Rho-like small GTPase (RLSG) activity, and demonstrate deficiency for mutants of pTSNS patients. We find that CDK20 regulates DLC1. Knockdown of MAGI2, DLC1, or CDK20 in cultured podocytes reduces migration rate. Treatment with dexamethasone abolishes RhoA activation by knockdown of DLC1 or CDK20 indicating that steroid treatment in patients with pTSNS and mutations in these genes is mediated by this RLSG module. Furthermore, we discover ITSN1 and ITSN2 as podocytic guanine nucleotide exchange factors for Cdc42. We generate Itsn2-L knockout mice that recapitulate the mild NS phenotype. We, thus, define a functional network of RhoA regulation, thereby revealing potential therapeutic targets.


Asunto(s)
Resistencia a Medicamentos/genética , Glucocorticoides/farmacología , Síndrome Nefrótico/tratamiento farmacológico , Mapas de Interacción de Proteínas/genética , Proteína de Unión al GTP rhoA/genética , Adulto , Animales , Niño , Preescolar , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Glucocorticoides/uso terapéutico , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mutación , Síndrome Nefrótico/genética , Linaje , Podocitos , ARN Interferente Pequeño/metabolismo , Resultado del Tratamiento , Secuenciación del Exoma , Proteína de Unión al GTP rhoA/metabolismo
5.
Clin J Am Soc Nephrol ; 13(1): 53-62, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-29127259

RESUMEN

BACKGROUND AND OBJECTIVES: Steroid-resistant nephrotic syndrome overwhelmingly progresses to ESRD. More than 30 monogenic genes have been identified to cause steroid-resistant nephrotic syndrome. We previously detected causative mutations using targeted panel sequencing in 30% of patients with steroid-resistant nephrotic syndrome. Panel sequencing has a number of limitations when compared with whole exome sequencing. We employed whole exome sequencing to detect monogenic causes of steroid-resistant nephrotic syndrome in an international cohort of 300 families. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Three hundred thirty-five individuals with steroid-resistant nephrotic syndrome from 300 families were recruited from April of 1998 to June of 2016. Age of onset was restricted to <25 years of age. Exome data were evaluated for 33 known monogenic steroid-resistant nephrotic syndrome genes. RESULTS: In 74 of 300 families (25%), we identified a causative mutation in one of 20 genes known to cause steroid-resistant nephrotic syndrome. In 11 families (3.7%), we detected a mutation in a gene that causes a phenocopy of steroid-resistant nephrotic syndrome. This is consistent with our previously published identification of mutations using a panel approach. We detected a causative mutation in a known steroid-resistant nephrotic syndrome gene in 38% of consanguineous families and in 13% of nonconsanguineous families, and 48% of children with congenital nephrotic syndrome. A total of 68 different mutations were detected in 20 of 33 steroid-resistant nephrotic syndrome genes. Fifteen of these mutations were novel. NPHS1, PLCE1, NPHS2, and SMARCAL1 were the most common genes in which we detected a mutation. In another 28% of families, we detected mutations in one or more candidate genes for steroid-resistant nephrotic syndrome. CONCLUSIONS: Whole exome sequencing is a sensitive approach toward diagnosis of monogenic causes of steroid-resistant nephrotic syndrome. A molecular genetic diagnosis of steroid-resistant nephrotic syndrome may have important consequences for the management of treatment and kidney transplantation in steroid-resistant nephrotic syndrome.


Asunto(s)
Análisis Mutacional de ADN/métodos , Secuenciación del Exoma , Marcadores Genéticos , Mutación , Síndrome Nefrótico/congénito , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Herencia , Humanos , Lactante , Masculino , Tasa de Mutación , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Síndrome Nefrótico/terapia , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Adulto Joven
6.
J Am Soc Nephrol ; 28(10): 3055-3065, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28566477

RESUMEN

We investigated the value of genetic, histopathologic, and early treatment response information in prognosing long-term renal outcome in children with primary steroid-resistant nephrotic syndrome. From the PodoNet Registry, we obtained longitudinal clinical information for 1354 patients (disease onset at >3 months and <20 years of age): 612 had documented responsiveness to intensified immunosuppression (IIS), 1155 had kidney biopsy results, and 212 had an established genetic diagnosis. We assessed risk factors for ESRD using multivariate Cox regression models. Complete and partial remission of proteinuria within 12 months of disease onset occurred in 24.5% and 16.5% of children, respectively, with the highest remission rates achieved with calcineurin inhibitor-based protocols. Ten-year ESRD-free survival rates were 43%, 94%, and 72% in children with IIS resistance, complete remission, and partial remission, respectively; 27% in children with a genetic diagnosis; and 79% and 52% in children with histopathologic findings of minimal change glomerulopathy and FSGS, respectively. Five-year ESRD-free survival rate was 21% for diffuse mesangial sclerosis. IIS responsiveness, presence of a genetic diagnosis, and FSGS or diffuse mesangial sclerosis on initial biopsy as well as age, serum albumin concentration, and CKD stage at onset affected ESRD risk. Our findings suggest that responsiveness to initial IIS and detection of a hereditary podocytopathy are prognostic indicators of favorable and poor long-term outcome, respectively, in children with steroid-resistant nephrotic syndrome. Children with multidrug-resistant sporadic disease show better renal survival than those with genetic disease. Furthermore, histopathologic findings may retain prognostic relevance when a genetic diagnosis is established.


Asunto(s)
Inmunosupresores/uso terapéutico , Fallo Renal Crónico/etiología , Síndrome Nefrótico/congénito , Adolescente , Niño , Preescolar , Estudios de Seguimiento , Humanos , Lactante , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/tratamiento farmacológico , Análisis de Supervivencia
7.
J Pediatr ; 177S: S156-S172, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27666265

RESUMEN

The health care system in Serbia is based on a network of public health institutions funded by the National Health Insurance and from the state budget. Access to public health institutions is free. Preventive and curative services are provided at the local level in primary health care centers. Over the past 5-7 years, the number of pediatricians in primary health care centers decreased because of reduced number of applicants for pediatric training, which endangers the maintenance of the traditional model of pediatric care. Secondary medical care is offered in pediatric departments of local and regional general hospitals or outpatient clinics, and in specialized hospitals for children or adults. Tertiary medical care is provided by inpatient or outpatient subspecialty services in 5 major university children's clinics. The health reforms undertaken in the recent 10 years have aimed at strengthening preventive health care and reducing the overall costs for pediatric care. Current initiatives of the Ministry of Health and national pediatric associations are aimed at reestablishing and strengthening the capacity of the primary pediatric health care model by increasing the number of physicians and developing new processes of care.


Asunto(s)
Servicios de Salud del Niño , Salud Infantil , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Serbia
8.
Nat Commun ; 7: 10822, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26905694

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) causes 15% of chronic kidney disease (CKD). Here we show that recessive mutations in FAT1 cause a distinct renal disease entity in four families with a combination of SRNS, tubular ectasia, haematuria and facultative neurological involvement. Loss of FAT1 results in decreased cell adhesion and migration in fibroblasts and podocytes and the decreased migration is partially reversed by a RAC1/CDC42 activator. Podocyte-specific deletion of Fat1 in mice induces abnormal glomerular filtration barrier development, leading to podocyte foot process effacement. Knockdown of Fat1 in renal tubular cells reduces migration, decreases active RAC1 and CDC42, and induces defects in lumen formation. Knockdown of fat1 in zebrafish causes pronephric cysts, which is partially rescued by RAC1/CDC42 activators, confirming a role of the two small GTPases in the pathogenesis. These findings provide new insights into the pathogenesis of SRNS and tubulopathy, linking FAT1 and RAC1/CDC42 to podocyte and tubular cell function.


Asunto(s)
Cadherinas/genética , Adhesión Celular/genética , Movimiento Celular/genética , Fibroblastos/metabolismo , Síndrome Nefrótico/congénito , Podocitos/metabolismo , Proteínas de Pez Cebra/genética , Animales , Dilatación Patológica/genética , Técnicas de Silenciamiento del Gen , Hematuria/genética , Humanos , Túbulos Renales/citología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Lisencefalia/genética , Ratones , Mutación , Síndrome Nefrótico/genética , Síndrome , Pez Cebra , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo
9.
Nat Genet ; 48(4): 457-65, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878725

RESUMEN

Nucleoporins are essential components of the nuclear pore complex (NPC). Only a few diseases have been attributed to NPC dysfunction. Steroid-resistant nephrotic syndrome (SRNS), a frequent cause of chronic kidney disease, is caused by dysfunction of glomerular podocytes. Here we identify in eight families with SRNS mutations in NUP93, its interaction partner NUP205 or XPO5 (encoding exportin 5) as hitherto unrecognized monogenic causes of SRNS. NUP93 mutations caused disrupted NPC assembly. NUP93 knockdown reduced the presence of NUP205 in the NPC, and, reciprocally, a NUP205 alteration abrogated NUP93 interaction. We demonstrate that NUP93 and exportin 5 interact with the signaling protein SMAD4 and that NUP93 mutations abrogated interaction with SMAD4. Notably, NUP93 mutations interfered with BMP7-induced SMAD transcriptional reporter activity. We hereby demonstrate that mutations of NUP genes cause a distinct renal disease and identify aberrant SMAD signaling as a new disease mechanism of SRNS, opening a potential new avenue for treatment.


Asunto(s)
Carioferinas/genética , Síndrome Nefrótico/genética , Proteínas de Complejo Poro Nuclear/genética , Edad de Inicio , Secuencia de Aminoácidos , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Resistencia a Medicamentos/genética , Femenino , Genes Recesivos , Estudios de Asociación Genética , Ligamiento Genético , Células HEK293 , Humanos , Lactante , Carioferinas/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Proteínas de Complejo Poro Nuclear/metabolismo , Estrés Oxidativo , Podocitos/fisiología , Análisis de Secuencia de ADN , Esteroides/farmacología , Esteroides/uso terapéutico , Xenopus laevis
10.
Acta Paediatr ; 105(1): e35-41, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26437121

RESUMEN

AIM: This studied reviewed renovascular hypertension (RVH) due to renal artery stenosis (RAS) in two Serbian paediatric centres from 2001 to 2013. METHODS: The patients' demographic data, underlying syndromes, blood pressure (BP), antihypertensive treatments and outcomes were reviewed. RESULTS: The incidence of RVH was 1.9 per million children per year during the study period, and there were 25 patients with RAS, aged 10.4 ± 5.2 years. At presentation, their mean blood pressure (BP) standard deviation scores were 6.9 ± 3.4 systolic and 5.2 ± 2.6 diastolic. BP loads on 24-hour ambulatory BP were 88 ± 14% systolic and 80 ± 29% diastolic. We found that 72% had fibromuscular dysplasia and 28% had underlying syndromes. RAS was unilateral in 64% and bilateral in 28%, and 8% had RAS of a single kidney. Antihypertensive treatment included antihypertensive drugs (100%), percutaneous transluminal angioplasty (92%), renal auto-transplantation (16%), surgical revascularisation (12%) and nephrectomy (12%). After 4.4 ± 3.6 years of follow-up, high BP was cured in 40% of the patients and 39.4% of the kidneys and improved in 48% (75.7%), with BP decreases of 20.3 ± 3.7% systolic and 16.3 ± 6.2% diastolic. CONCLUSION: Fibromuscular dysplasia was the most common cause of RVH in this study, and hypertension was cured or improved in 88% of the patients.


Asunto(s)
Displasia Fibromuscular/complicaciones , Hipertensión Renovascular/terapia , Obstrucción de la Arteria Renal/complicaciones , Adolescente , Antihipertensivos/uso terapéutico , Niño , Preescolar , Terapia Combinada , Femenino , Displasia Fibromuscular/diagnóstico , Estudios de Seguimiento , Humanos , Hipertensión Renovascular/diagnóstico , Hipertensión Renovascular/epidemiología , Hipertensión Renovascular/etiología , Trasplante de Riñón , Masculino , Nefrectomía , Obstrucción de la Arteria Renal/diagnóstico , Estudios Retrospectivos , Serbia/epidemiología , Resultado del Tratamiento
11.
Clin Immunol ; 161(2): 355-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26499378

RESUMEN

Schimke immuno-osseous dysplasia (SIOD) is an autosomal recessive, fatal childhood disorder associated with skeletal dysplasia, renal dysfunction, and T-cell immunodeficiency. This disease is linked to biallelic loss-of-function mutations of the SMARCAL1 gene. Although recurrent infection, due to T-cell deficiency, is a leading cause of morbidity and mortality, the etiology of the T-cell immunodeficiency is unclear. Here, we demonstrate that the T cells of SIOD patients have undetectable levels of protein and mRNA for the IL-7 receptor alpha chain (IL7Rα) and are unresponsive to stimulation with IL-7, indicating a loss of functional receptor. No pathogenic mutations were detected in the exons of IL7R in these patients; however, CpG sites in the IL7R promoter were hypermethylated in SIOD T cells. We propose therefore that the lack of IL7Rα expression, associated with hypermethylation of the IL7R promoter, in T cells and possibly their earlier progenitors, restricts T-cell development in SIOD patients.


Asunto(s)
Arteriosclerosis/genética , Síndromes de Inmunodeficiencia/genética , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Embolia Pulmonar/genética , Receptores de Interleucina-7/genética , Linfocitos T/metabolismo , Adolescente , Adulto , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Células Cultivadas , Niño , Preescolar , ADN Helicasas/genética , Metilación de ADN , Citometría de Flujo , Expresión Génica , Humanos , Inmunohistoquímica , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Interleucina-17/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Mutación , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Enfermedades de Inmunodeficiencia Primaria , Regiones Promotoras Genéticas/genética , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patología , Receptores de Interleucina-7/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Adulto Joven
12.
Pediatr Res ; 78(6): 609-17, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26309238

RESUMEN

BACKGROUND: Schimke immuno-osseous dysplasia (SIOD) is an autosomal recessive disorder caused by mutations in SMARCAL1. A frequent complication is arteriosclerosis associated with reduced elastin expression; however, the mechanism underlying the reduced elastin expression remains unknown. METHODS: Expression of transcriptional regulators of elastin (ELN) and microRNA (miRNA) regulators of ELN messenger RNA (mRNA), ELN promoter methylation, and ELN mRNA poly(A) tail length were assessed by quantitative RT-PCR, bisulfite Sanger sequencing, and the Poly(A) Tail Length Assay Kit, respectively, in unaffected developing human aortae and in an SIOD aorta. RESULTS: Comparing unaffected fetal and adult aortae, ELN precursor mRNA (pre-mRNA) levels remained nearly constant, whereas mRNA levels declined by ~10(2)-fold. This corresponded with a reduction in poly(A) tail length but not with changes in the other parameters. In contrast, compared to the unaffected fetal aortae, the SIOD aorta had 18-fold less ELN pre-mRNA and 10(4)-fold less mRNA. This corresponded with increased expression of miRNA regulators and shorter ELN mRNA poly(A) tail lengths but not with altered expression of ELN transcriptional regulators or ELN promoter methylation. CONCLUSION: Posttranscriptional mechanisms account for the reduction in ELN mRNA levels in unaffected aortae, whereas transcriptional and posttranscriptional mechanisms reduce elastin expression in SIOD aorta and predispose to arteriosclerosis.


Asunto(s)
Aorta/metabolismo , Arteriosclerosis/genética , Elastina/genética , Síndromes de Inmunodeficiencia/genética , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Embolia Pulmonar/genética , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Transcripción Genética , Adolescente , Adulto , Aorta/embriología , Aorta/patología , Arteriosclerosis/embriología , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Metilación de ADN , Regulación hacia Abajo , Elastina/metabolismo , Femenino , Edad Gestacional , Humanos , Síndromes de Inmunodeficiencia/embriología , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Síndrome Nefrótico/embriología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Osteocondrodisplasias/embriología , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Enfermedades de Inmunodeficiencia Primaria , Regiones Promotoras Genéticas , Embolia Pulmonar/embriología , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patología , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Hum Genet ; 134(8): 905-16, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26026792

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) account for 40-50% of chronic kidney disease that manifests in the first two decades of life. Thus far, 31 monogenic causes of isolated CAKUT have been described, explaining ~12% of cases. To identify additional CAKUT-causing genes, we performed whole-exome sequencing followed by a genetic burden analysis in 26 genetically unsolved families with CAKUT. We identified two heterozygous mutations in SRGAP1 in 2 unrelated families. SRGAP1 is a small GTPase-activating protein in the SLIT2-ROBO2 signaling pathway, which is essential for development of the metanephric kidney. We then examined the pathway-derived candidate gene SLIT2 for mutations in cohort of 749 individuals with CAKUT and we identified 3 unrelated individuals with heterozygous mutations. The clinical phenotypes of individuals with mutations in SLIT2 or SRGAP1 were cystic dysplastic kidneys, unilateral renal agenesis, and duplicated collecting system. We show that SRGAP1 is expressed in early mouse nephrogenic mesenchyme and that it is coexpressed with ROBO2 in SIX2-positive nephron progenitor cells of the cap mesenchyme in developing rat kidney. We demonstrate that the newly identified mutations in SRGAP1 lead to an augmented inhibition of RAC1 in cultured human embryonic kidney cells and that the SLIT2 mutations compromise the ability of the SLIT2 ligand to inhibit cell migration. Thus, we report on two novel candidate genes for causing monogenic isolated CAKUT in humans.


Asunto(s)
Proteínas Activadoras de GTPasa , Péptidos y Proteínas de Señalización Intercelular , Mutación , Proteínas del Tejido Nervioso , Receptores Inmunológicos , Transducción de Señal/genética , Anomalías Urogenitales , Reflujo Vesicoureteral , Animales , Exoma , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/metabolismo , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/metabolismo , Ratas , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Factores de Riesgo , Anomalías Urogenitales/embriología , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/embriología , Reflujo Vesicoureteral/genética
14.
Pediatr Nephrol ; 30(1): 79-90, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25129203

RESUMEN

BACKGROUND: A high prevalence of chronic kidney disease among children with focal segmental glomerulosclerosis (FSGS) leads to a permanent quest for good predictors of kidney dysfunction. Thus, we carried out a retrospective cohort study in order to examine known clinical and morphological predictors of adverse outcome, as well as to investigate glomerular nestin expression as a potential new early predictor of kidney dysfunction in children with FSGS. Relationships between nestin expression and clinical and morphological findings were also investigated. METHODS: Among 649 renal biopsy samples, obtained from two children's hospitals, FSGS was diagnosed in 60 children. Thirty-eight patients, who met the criteria for this study, were followed up for 9.0 ± 5.2 years. Using Kaplan-Meier and Cox's regression analysis, potential clinical and morphological predictors were applied in two models of prediction: after disease onset and after the biopsy. RESULTS: The present study revealed the following significant predictors of kidney dysfunction: patients' ages at disease onset, as well as age at biopsy, resistance to corticosteroid treatment, serum creatinine level, urine protein/creatinine ratio, vascular involvement, tubular atrophy, interstitial fibrosis, and decreased glomerular nestin expression. CONCLUSIONS: The most important finding of our study is that nestin can be used as a potential new early morphological predictor of kidney dysfunction in childhood onset of FSGS, since nestin has been obviously decreased in both sclerotic and normal glomeruli seen by light microscopy.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomérulos Renales/metabolismo , Nestina/análisis , Adolescente , Biomarcadores/análisis , Niño , Preescolar , Estudios de Cohortes , Femenino , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Humanos , Lactante , Fallo Renal Crónico/epidemiología , Masculino , Estudios Retrospectivos , Adulto Joven
15.
Vojnosanit Pregl ; 71(4): 395-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24783421

RESUMEN

INTRODUCTION: The group of autosomal dominant disorders - Epstein syndrome, Sebastian syndrome, Fechthner syndrome and May-Hegglin anomaly - are characterised by thrombocytopenia with giant platelets, inclusion bodies in granulocytes and variable levels of deafness, disturbances of vision and renal function impairment. A common genetic background of these disorders are mutations in MYH9 gene, coding for the nonmuscle myosin heavy chain IIA. Differential diagnosis is important for the adequate treatment strategy. The aim of this case report was to present a patient with MYH9 disorder in Serbia. CASE REPORT: A 16-year-old boy was referred to our hospital with the diagnosis of resistant immune thrombocytopenia for splenectomy. Thrombocytopenia was incidentally discovered at the age of five. The treatment with corticosteroids on several occasions was unsuccessful. Although the platelet count was below 10 x 10(9)/L, there were no bleeding symptoms. Besides thrombocytopenia with giant platelets, on admission the patient also suffered sensorineuronal hearing loss and proteinuria. The diagnosis was confirmed with immunofluorescence and genetic analyses. CONCLUSION: Early recognition of MYH9-related diseases is essential to avoid unnecessary and potentially harmful treatments for misdiagnosed immune thrombocytopenia, and also for timely and proper therapy in attempt to delay end-stage renal failure and improve quality of life.


Asunto(s)
Pérdida Auditiva Sensorineural/diagnóstico , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Nefritis/etiología , Trombocitopenia/congénito , Adolescente , Diagnóstico Diferencial , Técnica del Anticuerpo Fluorescente , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Mutación , Nefritis/congénito , Recuento de Plaquetas , Serbia , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/fisiopatología
16.
J Nephrol ; 27(5): 537-44, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24756972

RESUMEN

BACKGROUND: Growth retardation is one of the most visible comorbid conditions of chronic kidney disease (CKD) in children. To our knowledge, published data on longitudinal follow-up of growth in pediatric patients with CKD is lacking from the region of South-East Europe. Herein we report the results from the Serbian Pediatric Registry of Chronic Kidney Disease. METHODS: The data reported in the present prospective analysis were collected between 2000 and 2012. A total of 324 children with CKD were enrolled in the registry. RESULTS: Prevalence of growth failure at registry entry was 29.3 %. Mean height standard deviation scores (HtSDS) in children with stunting and those with normal stature were -3.00 [95 % confidence interval (CI) -3.21 to -2.79] and -0.08 (95 % CI -0.22 to 0.05) (p < 0.001), respectively. Children with hereditary nephropathy had worse growth at registration (-1.51; 95 % CI -1.97 to -1.04, p = 0.008). Those with CKD stages 4 and 5 before registration had more chance to have short stature at registration than those with CKD stages 2 and 3 [odds ratio (OR) = 0.458, CI 0.268-0.782, p = 0.004]. Dialysis was an independent negative predictor for maintaining optimal stature during the follow-up period (OR = 0.324, CI = 0.199-0.529, p < 0.001), while transplantation was an independent positive predictor for improvement of small stature during follow-up (OR = 3.706, CI = 1.785-7.696, p < 0.001). CONCLUSION: Growth failure remains a significant problem in children with CKD, being worst in patients with hereditary renal disease. Growth is not improved by standard dialysis, but transplantation has a positive impact on growth in children.


Asunto(s)
Estatura , Trastornos del Crecimiento/epidemiología , Insuficiencia Renal Crónica/epidemiología , Adolescente , Factores de Edad , Distribución de Chi-Cuadrado , Niño , Preescolar , Comorbilidad , Femenino , Estudios de Seguimiento , Trastornos del Crecimiento/diagnóstico , Trastornos del Crecimiento/fisiopatología , Humanos , Lactante , Recién Nacido , Trasplante de Riñón , Modelos Logísticos , Masculino , Análisis Multivariante , Oportunidad Relativa , Prevalencia , Estudios Prospectivos , Sistema de Registros , Diálisis Renal , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/terapia , Factores de Riesgo , Serbia/epidemiología , Factores de Tiempo , Adulto Joven
17.
J Am Soc Nephrol ; 25(9): 1917-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24700879

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately 40% of children with ESRD in the United States. Hitherto, mutations in 23 genes have been described as causing autosomal dominant isolated CAKUT in humans. However, >90% of cases of isolated CAKUT still remain without a molecular diagnosis. Here, we hypothesized that genes mutated in recessive mouse models with the specific CAKUT phenotype of unilateral renal agenesis may also be mutated in humans with isolated CAKUT. We applied next-generation sequencing technology for targeted exon sequencing of 12 recessive murine candidate genes in 574 individuals with isolated CAKUT from 590 families. In 15 of 590 families, we identified recessive mutations in the genes FRAS1, FREM2, GRIP1, FREM1, ITGA8, and GREM1, all of which function in the interaction of the ureteric bud and the metanephric mesenchyme. We show that isolated CAKUT may be caused partially by mutations in recessive genes. Our results also indicate that biallelic missense mutations in the Fraser/MOTA/BNAR spectrum genes cause isolated CAKUT, whereas truncating mutations are found in the multiorgan form of Fraser syndrome. The newly identified recessive biallelic mutations in these six genes represent the molecular cause of isolated CAKUT in 2.5% of the 590 affected families in this study.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Matriz Extracelular/genética , Síndrome de Fraser/genética , Cadenas alfa de Integrinas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Riñón/anomalías , Mutación , Proteínas del Tejido Nervioso/genética , Receptores de Interleucina/genética , Sistema Urinario/anomalías , Reflujo Vesicoureteral/genética , Animales , Anomalías Congénitas/genética , Modelos Animales de Enfermedad , Femenino , Genes Recesivos , Humanos , Enfermedades Renales/congénito , Enfermedades Renales/genética , Masculino , Ratones , Ratones Mutantes , Anomalías Urogenitales
18.
Kidney Int ; 85(6): 1429-33, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24429398

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease. CAKUT can be caused by monogenic mutations; however, data are lacking on their frequency. Genetic diagnosis has been hampered by genetic heterogeneity and lack of genotype-phenotype correlation. To determine the percentage of cases with CAKUT that can be explained by mutations in known CAKUT genes, we analyzed the coding exons of the 17 known dominant CAKUT-causing genes in a cohort of 749 individuals from 650 families with CAKUT. The most common phenotypes in this CAKUT cohort were vesicoureteral reflux in 288 patients, renal hypodysplasia in 120 patients, and unilateral renal agenesis in 90 patients. We identified 37 different heterozygous mutations (33 novel) in 12 of the 17 known genes in 47 patients from 41 of the 650 families (6.3%). These mutations include (number of families): BMP7 (1), CDC5L (1), CHD1L (5), EYA1 (3), GATA3 (2), HNF1B (6), PAX2 (5), RET (3), ROBO2 (4), SALL1 (9), SIX2 (1), and SIX5 (1). Furthermore, several mutations previously reported to be disease-causing are most likely benign variants. Thus, in a large cohort over 6% of families with isolated CAKUT are caused by a mutation in 12 of 17 dominant CAKUT genes. Our report represents one of the most in-depth diagnostic studies of monogenic causes of isolated CAKUT in children.


Asunto(s)
Genes Dominantes , Mutación , Reflujo Vesicoureteral/genética , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Herencia , Heterocigoto , Humanos , Masculino , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Anomalías Urogenitales
19.
Kidney Int ; 85(5): 1169-78, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24402088

RESUMEN

WT1 mutations cause a wide spectrum of renal and extrarenal manifestations. Here we evaluated disease prevalence, phenotype spectrum, and genotype-phenotype correlations of 61 patients with WT1-related steroid-resistant nephrotic syndrome relative to 700 WT1-negative patients, all with steroid-resistant nephrotic syndrome. WT1 patients more frequently presented with chronic kidney disease and hypertension at diagnosis and exhibited more rapid disease progression. Focal segmental glomerulosclerosis was equally prevalent in both cohorts, but diffuse mesangial sclerosis was largely specific for WT1 disease and was present in 34% of cases. Sex reversal and/or urogenital abnormalities (52%), Wilms tumor (38%), and gonadoblastoma (5%) were almost exclusive to WT1 disease. Missense substitutions affecting DNA-binding residues were associated with diffuse mesangial sclerosis (74%), early steroid-resistant nephrotic syndrome onset, and rapid progression to ESRD. Truncating mutations conferred the highest Wilms tumor risk (78%) but typically late-onset steroid-resistant nephrotic syndrome. Intronic (KTS) mutations were most likely to present as isolated steroid-resistant nephrotic syndrome (37%) with a median onset at an age of 4.5 years, focal segmental glomerulosclerosis on biopsy, and slow progression (median ESRD age 13.6 years). Thus, there is a wide range of expressivity, solid genotype-phenotype associations, and a high risk and significance of extrarenal complications in WT1-associated nephropathy. We suggest that all children with steroid-resistant nephrotic syndrome undergo WT1 gene screening.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Mutación , Síndrome Nefrótico/congénito , Insuficiencia Renal Crónica/genética , Proteínas WT1/genética , Edad de Inicio , Niño , Preescolar , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Glomeruloesclerosis Focal y Segmentaria/epidemiología , Glomeruloesclerosis Focal y Segmentaria/terapia , Humanos , Incidencia , Lactante , Masculino , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Síndrome Nefrótico/terapia , Fenotipo , Prevalencia , Pronóstico , Sistema de Registros , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/terapia , Factores de Riesgo , Factores de Tiempo
20.
Kidney Int ; 85(6): 1310-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24152966

RESUMEN

Congenital abnormalities of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease and they are the most frequent cause of end-stage renal disease in children in the US. However, its genetic etiology remains mostly elusive. VACTERL association is a rare disorder that involves congenital abnormalities in multiple organs including the kidney and urinary tract in up to 60% of the cases. By homozygosity mapping and whole-exome resequencing combined with high-throughput mutation analysis by array-based multiplex PCR and next-generation sequencing, we identified recessive mutations in the gene TNF receptor-associated protein 1 (TRAP1) in two families with isolated CAKUT and three families with VACTERL association. TRAP1 is a heat-shock protein 90-related mitochondrial chaperone possibly involved in antiapoptotic and endoplasmic reticulum stress signaling. Trap1 is expressed in renal epithelia of developing mouse kidney E13.5 and in the kidney of adult rats, most prominently in proximal tubules and in thick medullary ascending limbs of Henle's loop. Thus, we identified mutations in TRAP1 as highly likely causing CAKUT or VACTERL association with CAKUT.


Asunto(s)
Canal Anal/anomalías , Análisis Mutacional de ADN , Esófago/anomalías , Exosomas , Pruebas Genéticas , Proteínas HSP90 de Choque Térmico , Cardiopatías Congénitas/genética , Riñón/anomalías , Deformidades Congénitas de las Extremidades/genética , Mutación , Columna Vertebral/anomalías , Tráquea/anomalías , Reflujo Vesicoureteral/genética , Factores de Edad , Animales , Análisis Mutacional de ADN/métodos , Europa (Continente) , Femenino , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Edad Gestacional , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Cardiopatías Congénitas/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Riñón/embriología , Riñón/metabolismo , Deformidades Congénitas de las Extremidades/diagnóstico , Masculino , Ratones , Reacción en Cadena de la Polimerasa Multiplex , Linaje , Valor Predictivo de las Pruebas , Factores de Riesgo , Estados Unidos , Anomalías Urogenitales , Reflujo Vesicoureteral/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA