Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Exp Eye Res ; 231: 109499, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37169279

RESUMEN

Fuchs Endothelial Corneal Dystrophy (FECD), a late-onset oxidative stress disorder, is the most common cause of corneal endothelial degeneration and is genetically associated with CTG repeat expansion in Transcription Factor 4 (TCF4). We previously reported accumulation of nuclear (nDNA) and mitochondrial (mtDNA) damage in FECD. Specifically, mtDNA damage was a prominent finding in development of disease in the ultraviolet-A (UVA) induced FECD mouse model. We hypothesize that an aberrant DNA repair may contribute to the increased DNA damage seen in FECD. We analyzed differential expression profiles of 84 DNA repair genes by real-time PCR arrays using Human DNA Repair RT-Profiler plates using cDNA extracted from Descemet's membrane-corneal endothelium (DM-CE) obtained from FECD patients with expanded (>40) or non-expanded (<40) intronic CTG repeats in TCF4 gene and from age-matched normal donors. Change in mRNA expression of <0.5- or >2.0-fold in FECD relative to normal was set as cutoff for down- or upregulation. Downregulated mitochondrial genes were further validated using the UVA-based mouse model of FECD. FECD specimens exhibited downregulation of 9 genes and upregulation of 8 genes belonging to the four major DNA repair pathways, namely, base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), and double strand break (DSB) repair, compared to normal donors. MMR gene MSH2 and BER gene POLB were preferentially upregulated in expanded FECD. BER genes LIG3 and NEIL2, DSB repair genes PARP3 and TOP3A, NER gene XPC, and unclassified pathway gene TREX1, were downregulated in both expanded and non-expanded FECD. MtDNA repair genes, Lig3, Neil2, and Top3a, were also downregulated in the UVA-based mouse model of FECD. Our findings identify impaired DNA repair pathways that may play an important role in DNA damage due to oxidative stress as well as genetic predisposition noted in FECD.


Asunto(s)
ADN Glicosilasas , Distrofia Endotelial de Fuchs , Animales , Ratones , Humanos , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Endotelio Corneal/metabolismo , Predisposición Genética a la Enfermedad , Reparación del ADN/genética , ADN Mitocondrial/genética , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo
2.
J Pak Med Assoc ; 72(1): 22-26, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35099432

RESUMEN

OBJECTIVE: To assess the relative efficacy of flaxseed and fish oils in the management of rheumatoid arthritis. METHODS: The comparative study was conducted in the outpatient department of Rheumatology at Fatima Memorial Hospital, Shadman, Lahore, Pakistan, from July to December 2017, and comprised rheumatoid arthritis patients who were divided into group A receiving 3g/day of flaxseed oil and group B receiving 3g/day of fish oil for 90 days. Blood samples were taken at baseline and post-intervention to note the difference on biochemical parameters. Data was analysed using SPSS 21. RESULTS: Of the 60 patients, there were 30(50%) in each of the two groups. Overall, there were 8(13.3%) males and 52(86.7%) females. Both groups showed significant change in all biochemical parameters compared to baseline values (p<0.05). Intergroup comparison showed that flaxseed oil treatment was significantly more effective than fish oil treatment (p<0.05). CONCLUSIONS: While both forms of intervention were found to be effective, flaxseed oil was significantly more effective.


Asunto(s)
Artritis Reumatoide , Lino , Adulto , Artritis Reumatoide/tratamiento farmacológico , Femenino , Aceites de Pescado/uso terapéutico , Humanos , Aceite de Linaza , Masculino , Pakistán
3.
Pak J Med Sci ; 38(8): 2307-2312, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36415236

RESUMEN

Objectives: The study was aimed at evaluating the association between junk food consumption and BMI of adolescent girls along with the menstrual abnormalities and to compare it with controls. Methods: A cross-sectional study was conducted among 200 girls between 13 - 19 years of age at Bahria International Hospital, Lahore based on self-administered questionnaire from July 2021 to September 2021. The total subjects were divided in two groups Viz; Group-A which comprised of 100 girls with menstrual abnormalities and Group-B included 100 girls without menstrual problem (control group). The data recorded on the questionnaire about the demographic profile, anthropometric measurements, menstrual cycle characteristics, and dietary habits was subjected to statistical analysis using SPSS version 20 and Chi-Square was used to test quantitative significance between the two groups. Results: The mean age of participants was 17.02±1.76 years. It was observed that 40% girls had irregular menstrual cycle, 56% girls were suffering from dysmenorrhea and almost all girls of Group-I were suffering from premenstrual dysfunctions. The current study found a non-significant difference between two groups with regard to body mass index (P≥0.05). Significant difference was observed between two groups (P ≤ 0.05) as junk food consumption was high in Group-A as compared to Group-B. However, no significant difference was found between Group-A and B in relation to the consumption of salty snacks and frozen meat items (P≥0.05). Conclusion: The results suggested that junk food consumption affects menstrual cycle negatively however more studies are needed to confirm the association of BMI, consumption of salty snacks and frozen meat items with menstrual abnormalities.

4.
J Am Soc Nephrol ; 30(2): 201-215, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30655312

RESUMEN

BACKGROUND: Whole-exome sequencing (WES) finds a CKD-related mutation in approximately 20% of patients presenting with CKD before 25 years of age. Although provision of a molecular diagnosis could have important implications for clinical management, evidence is lacking on the diagnostic yield and clinical utility of WES for pediatric renal transplant recipients. METHODS: To determine the diagnostic yield of WES in pediatric kidney transplant recipients, we recruited 104 patients who had received a transplant at Boston Children's Hospital from 2007 through 2017, performed WES, and analyzed results for likely deleterious variants in approximately 400 genes known to cause CKD. RESULTS: By WES, we identified a genetic cause of CKD in 34 out of 104 (32.7%) transplant recipients. The likelihood of detecting a molecular genetic diagnosis was highest for patients with urinary stone disease (three out of three individuals), followed by renal cystic ciliopathies (seven out of nine individuals), steroid-resistant nephrotic syndrome (nine out of 21 individuals), congenital anomalies of the kidney and urinary tract (ten out of 55 individuals), and chronic glomerulonephritis (one out of seven individuals). WES also yielded a molecular diagnosis for four out of nine individuals with ESRD of unknown etiology. The WES-related molecular genetic diagnosis had implications for clinical care for five patients. CONCLUSIONS: Nearly one third of pediatric renal transplant recipients had a genetic cause of their kidney disease identified by WES. Knowledge of this genetic information can help guide management of both transplant patients and potential living related donors.


Asunto(s)
Secuenciación del Exoma/métodos , Trasplante de Riñón/métodos , Medicina de Precisión/métodos , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/cirugía , Adolescente , Boston , Niño , Preescolar , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Pruebas Genéticas/métodos , Rechazo de Injerto , Supervivencia de Injerto , Hospitales Pediátricos , Humanos , Trasplante de Riñón/efectos adversos , Masculino , Pronóstico , Insuficiencia Renal Crónica/fisiopatología , Estudios Retrospectivos , Medición de Riesgo , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Receptores de Trasplantes/estadística & datos numéricos , Resultado del Tratamiento
5.
Kidney Int ; 95(5): 1079-1090, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31010479

RESUMEN

Recently, recessive mutations of MAGI2 were identified as a cause of steroid-resistant nephrotic syndrome (SRNS) in humans and mice. To further delineate the pathogenesis of MAGI2 loss of function, we generated stable knockout lines for the two zebrafish orthologues magi2a and magi2b by CRISPR/Cas9. We also developed a novel assay for the direct detection of proteinuria in zebrafish independent of transgenic background. Whereas knockout of magi2b did not yield a nephrotic syndrome phenotype, magi2a-/- larvae developed ascites, periorbital edema, and proteinuria, as indicated by increased excretion of low molecular weight protein. Electron microscopy demonstrated extensive podocyte foot process effacement. As in human SRNS, we observed genotype/phenotype correlation, with edema onset occurring earlier in zebrafish with truncating alleles (5-6 days post fertilization) versus hypomorphic alleles (19-20 days post fertilization). Paradoxically, corticosteroid treatment exacerbated the phenotype, with earlier onset of edema. In contrast, treatment with cyclosporine A or tacrolimus had no significant effect. Although RhoA signaling has been implicated as a downstream mediator of MAGI2 activity, targeting of the RhoA pathway did not modify the nephrotic syndrome phenotype. In the first CRISPR/Cas9 zebrafish knockout model of SRNS, we found that corticosteroids may have a paradoxical effect in the setting of specific genetic mutations.


Asunto(s)
Glucocorticoides/farmacología , Inmunosupresores/farmacología , Proteínas de la Membrana/genética , Síndrome Nefrótico/tratamiento farmacológico , Proteinuria/tratamiento farmacológico , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Ciclosporina/farmacología , Ciclosporina/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Medicamentos , Técnicas de Inactivación de Genes , Glucocorticoides/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Proteínas de Unión al GTP Monoméricas/metabolismo , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/genética , Proteinuria/patología , Transducción de Señal/efectos de los fármacos , Tacrolimus/farmacología , Tacrolimus/uso terapéutico , Resultado del Tratamiento , Pez Cebra , Proteínas de Pez Cebra/metabolismo
6.
Kidney Int ; 96(3): 642-655, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31171376

RESUMEN

The essential role of membrane associated guanylate kinase 2 (MAGI2) in podocytes is indicated by the phenotypes of severe glomerulosclerosis of both MAGI2 knockout mice and in patients with congenital nephrotic syndrome (CNS) caused by mutations in MAGI2. Here, we show that MAGI2 forms a complex with the Rap1 guanine nucleotide exchange factor, RapGEF2, and that this complex is lost when expressing MAGI2 CNS variants. Co-expression of RapGEF2 with wild-type MAGI2, but not MAGI2 CNS variants, enhanced activation of the small GTPase Rap1, a central signaling node in podocytes. In mice, podocyte-specific RapGEF2 deletion resulted in spontaneous glomerulosclerosis, with qualitative glomerular features comparable to MAGI2 knockout mice. Knockdown of RapGEF2 or MAGI2 in human podocytes caused similar reductions in levels of Rap1 activation and Rap1-mediated downstream signaling. Furthermore, human podocytes expressing MAGI2 CNS variants show severe abnormalities of cellular morphology and dramatic loss of actin cytoskeletal organization, features completely rescued by pharmacological activation of Rap1 via a non-MAGI2 dependent upstream pathway. Finally, immunostaining of kidney sections from patients with congenital nephrotic syndrome and MAGI2 mutations showed reduced podocyte Rap1-mediated signaling. Thus, MAGI2-RapGEF2-Rap1 signaling is essential for normal podocyte function. Hence, disruption of this pathway is an important cause of the renal phenotype induced by MAGI2 CNS mutations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanilato-Quinasas/genética , Síndrome Nefrótico/genética , Proteínas del Tejido Nervioso/metabolismo , Podocitos/patología , Proteínas de Unión a Telómeros/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Guanilato-Quinasas/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación , Síndrome Nefrótico/patología , Complejo Shelterina , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Unión a Telómeros/agonistas , Proteínas de Unión al GTP rap1/metabolismo
7.
Biochem Biophys Res Commun ; 517(3): 421-426, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31378371

RESUMEN

Antithrombin (AT3) is one of the most important inhibitors of blood coagulation proteases that belong to the serpin family of protease inhibitors. In this study, a novel alternatively spliced isoform of AT3 was identified, both at transcript and protein level. This novel transcript contains an additional region in the continuation of exon 3b that was included in the transcript due to use of an alternate 5' splice site. The existence of the novel transcript was confirmed in human brain and liver through RT-PCR. An analysis of the complete transcript indicated that the native reactive centre loop (RCL) of AT3 is maintained; however the novel amino acid sequence projects out as an additional loop as evident from MD simulation studies. A unique amino acid sequence present in the novel isoform was used for the development of polyclonal antibody. The expression of novel isoform was confirmed in human brain and liver tissue using Western blot analysis. Interestingly an alignment of RCL like domain with other inhibitory serpins showed significant similarity with the neuroserpin RCL. To the best of our knowledge, this is the first evidence of alternatively spliced AT3 sequence containing an additional loop and could have physiological relevance.


Asunto(s)
Empalme Alternativo , Antitrombina III/química , Heparina/química , Neuropéptidos/química , Serpinas/química , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Anticuerpos/aislamiento & purificación , Antitrombina III/genética , Antitrombina III/metabolismo , Secuencia de Bases , Sitios de Unión , Encéfalo/metabolismo , Expresión Génica , Heparina/metabolismo , Humanos , Hígado/metabolismo , Simulación de Dinámica Molecular , Neuropéptidos/genética , Neuropéptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Conejos , Serpinas/genética , Serpinas/metabolismo , Neuroserpina
8.
Nephrol Dial Transplant ; 34(3): 485-493, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29534211

RESUMEN

BACKGROUND: Nephrotic syndrome (NS), a chronic kidney disease, is characterized by significant loss of protein in the urine causing hypoalbuminemia and edema. In general, ∼15% of childhood-onset cases do not respond to steroid therapy and are classified as steroid-resistant NS (SRNS). In ∼30% of cases with SRNS, a causative mutation can be detected in one of 44 monogenic SRNS genes. The gene LAMA5 encodes laminin-α5, an essential component of the glomerular basement membrane. Mice with a hypomorphic mutation in the orthologous gene Lama5 develop proteinuria and hematuria. METHODS: To identify additional monogenic causes of NS, we performed whole exome sequencing in 300 families with pediatric NS. In consanguineous families we applied homozygosity mapping to identify genomic candidate loci for the underlying recessive mutation. RESULTS: In three families, in whom mutations in known NS genes were excluded, but in whom a recessive, monogenic cause of NS was strongly suspected based on pedigree information, we identified homozygous variants of unknown significance (VUS) in the gene LAMA5. While all affected individuals had nonsyndromic NS with an early onset of disease, their clinical outcome and response to immunosuppressive therapy differed notably. CONCLUSION: We here identify recessive VUS in the gene LAMA5 in patients with partially treatment-responsive NS. More data will be needed to determine the impact of these VUS in disease management. However, familial occurrence of disease, data from genetic mapping and a mouse model that recapitulates the NS phenotypes suggest that these genetic variants may be inherited factors that contribute to the development of NS in pediatric patients.


Asunto(s)
Secuenciación del Exoma/métodos , Inmunosupresores/uso terapéutico , Laminina/genética , Mutación , Síndrome Nefrótico/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología , Linaje , Fenotipo , Pronóstico , Adulto Joven
9.
Nephrol Dial Transplant ; 34(3): 474-485, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295827

RESUMEN

BACKGROUND: Alport syndrome (AS) and atypical hemolytic-uremic syndrome (aHUS) are rare forms of chronic kidney disease (CKD) that can lead to a severe decline of renal function. Steroid-resistant nephrotic syndrome (SRNS) is more common than AS and aHUS and causes 10% of childhood-onset CKD. In recent years, multiple monogenic causes of AS, aHUS and SRNS have been identified, but their relative prevalence has yet to be studied together in a typical pediatric cohort of children with proteinuria and hematuria. We hypothesized that identification of causative mutations by whole exome sequencing (WES) in known monogenic nephritis and nephrosis genes would allow distinguishing nephritis from nephrosis in a typical pediatric group of patients with both proteinuria and hematuria at any level. METHODS: We therefore conducted an exon sequencing (WES) analysis for 11 AS, aHUS and thrombotic thrombocytopenic purpura-causing genes in an international cohort of 371 patients from 362 families presenting with both proteinuria and hematuria before age 25 years. In parallel, we conducted either WES or high-throughput exon sequencing for 23 SRNS-causing genes in all patients. RESULTS: We detected pathogenic mutations in 18 of the 34 genes analyzed, leading to a molecular diagnosis in 14.1% of families (51 of 362). Disease-causing mutations were detected in 3 AS-causing genes (4.7%), 3 aHUS-causing genes (1.4%) and 12 NS-causing genes (8.0%). We observed a much higher mutation detection rate for monogenic forms of CKD in consanguineous families (35.7% versus 10.1%). CONCLUSIONS: We present the first estimate of relative frequency of inherited AS, aHUS and NS in a typical pediatric cohort with proteinuria and hematuria. Important therapeutic and preventative measures may result from mutational analysis in individuals with proteinuria and hematuria.


Asunto(s)
Secuenciación del Exoma/métodos , Marcadores Genéticos , Mutación , Nefritis/diagnóstico , Nefritis/genética , Nefrosis/diagnóstico , Nefrosis/genética , Adolescente , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/genética , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/genética , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Pronóstico
10.
J Am Soc Nephrol ; 29(8): 2123-2138, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29959197

RESUMEN

BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS. METHODS: To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes. RESULTS: We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog. CONCLUSIONS: Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Síndrome Nefrótico/genética , Podocitos/metabolismo , Proteínas de Unión al GTP rab5/genética , Animales , Movimiento Celular/genética , Células Cultivadas , Estudios de Cohortes , Progresión de la Enfermedad , Drosophila melanogaster , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Tamizaje Masivo/métodos , Mutación Missense , Síndrome Nefrótico/patología , Linaje , Proteínas de Unión a Fosfato , Podocitos/patología , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Secuenciación del Exoma
11.
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
12.
Hum Mutat ; 39(3): 406-414, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29194833

RESUMEN

Mutations in COQ8B cause steroid-resistant nephrotic syndrome with variable neurological involvement. In yeast, COQ8 encodes a protein required for coenzyme Q (CoQ) biosynthesis, whose precise role is not clear. Humans harbor two paralog genes: COQ8A and COQ8B (previously termed ADCK3 and ADCK4). We have found that COQ8B is a mitochondrial matrix protein peripherally associated with the inner membrane. COQ8B can complement a ΔCOQ8 yeast strain when its mitochondrial targeting sequence (MTS) is replaced by a yeast MTS. This model was employed to validate COQ8B mutations, and to establish genotype-phenotype correlations. All mutations affected respiratory growth, but there was no correlation between mutation type and the severity of the phenotype. In fact, contrary to the case of COQ2, where residual CoQ biosynthesis correlates with clinical severity, patients harboring hypomorphic COQ8B alleles did not display a different phenotype compared with those with null mutations. These data also suggest that the system is redundant, and that other proteins (probably COQ8A) may partially compensate for the absence of COQ8B. Finally, a COQ8B polymorphism, present in 50% of the European population (NM_024876.3:c.521A > G, p.His174Arg), affects stability of the protein and could represent a risk factor for secondary CoQ deficiencies or for other complex traits.


Asunto(s)
Resistencia a Medicamentos/genética , Mutación/genética , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/genética , Proteínas Quinasas/genética , Esteroides/uso terapéutico , Adolescente , Adulto , Niño , Preescolar , Estabilidad de Enzimas , Prueba de Complementación Genética , Humanos , Lactante , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Polimorfismo Genético , Saccharomyces cerevisiae/metabolismo , Adulto Joven
13.
Am J Hum Genet ; 96(1): 153-61, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25557779

RESUMEN

Nephrotic syndrome (NS), the association of gross proteinuria, hypoalbuminaemia, edema, and hyperlipidemia, can be clinically divided into steroid-sensitive (SSNS) and steroid-resistant (SRNS) forms. SRNS regularly progresses to end-stage renal failure. By homozygosity mapping and whole exome sequencing, we here identify recessive mutations in Crumbs homolog 2 (CRB2) in four different families affected by SRNS. Previously, we established a requirement for zebrafish crb2b, a conserved regulator of epithelial polarity, in podocyte morphogenesis. By characterization of a loss-of-function mutation in zebrafish crb2b, we now show that zebrafish crb2b is required for podocyte foot process arborization, slit diaphragm formation, and proper nephrin trafficking. Furthermore, by complementation experiments in zebrafish, we demonstrate that CRB2 mutations result in loss of function and therefore constitute causative mutations leading to NS in humans. These results implicate defects in podocyte apico-basal polarity in the pathogenesis of NS.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Síndrome Nefrótico/genética , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Niño , Preescolar , Mapeo Cromosómico , Exoma , Genes Recesivos , Homocigoto , Humanos , Lactante , Fallo Renal Crónico/etiología , Fallo Renal Crónico/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutación , Síndrome Nefrótico/complicaciones , Podocitos , Ratas , Pez Cebra/genética
14.
Am J Med Genet A ; 176(11): 2460-2465, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30079490

RESUMEN

Galloway-Mowat syndrome (GAMOS) is a phenotypically heterogeneous disorder characterized by neurodevelopmental defects combined with renal-glomerular disease, manifesting with proteinuria. To identify additional monogenic disease causes, we here performed whole exome sequencing (WES), linkage analysis, and homozygosity mapping in three affected siblings of an Indian family with GAMOS. Applying established criteria for variant filtering, we identify a novel homozygous splice site mutation in the gene WDR4 as the likely disease-causing mutation in this family. In line with previous reports, we observe growth deficiency, microcephaly, developmental delay, and intellectual disability as phenotypic features resulting from WDR4 mutations. However, the newly identified allele additionally gives rise to proteinuria and nephrotic syndrome, a phenotype that was never reported in patients with WDR4 mutations. Our data thus expand the phenotypic spectrum of WDR4 mutations by demonstrating that, depending on the specific mutated allele, a renal phenotype may be present. This finding suggests that GAMOS may occupy a phenotypic spectrum with other microcephalic diseases. Furthermore, WDR4 is an additional example of a gene that encodes a tRNA modifying enzyme and gives rise to GAMOS, if mutated. Our findings thereby support the recent observation that, like neurons, podocytes of the renal glomerulus are particularly vulnerable to cellular defects resulting from altered tRNA modifications.


Asunto(s)
Proteínas de Unión al GTP/genética , Hernia Hiatal/genética , Microcefalia/genética , Mutación , Nefrosis/genética , Adolescente , Niño , Preescolar , Genes Recesivos , Humanos , Secuenciación del Exoma
15.
Pediatr Nephrol ; 33(2): 305-314, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28921387

RESUMEN

BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is the second most frequent cause of end-stage renal disease (ESRD) among patients manifesting at under 25 years of age. We performed mutation analysis using a high-throughput PCR-based microfluidic technology in 24 single-gene causes of SRNS in a cohort of 72 families, who presented with SRNS before the age of 25 years. METHODS: Within an 18-month interval, we obtained DNA samples, pedigree information, and clinical information from 77 consecutive children with SRNS from 72 different families seen at Boston Children's Hospital (BCH). Mutation analysis was completed by combining high-throughput multiplex PCR with next-generation sequencing. We analyzed the sequences of 18 recessive and 6 dominant genes of SRNS in all 72 families for disease-causing variants. RESULTS: We identified the disease-causing mutation in 8 out of 72 (11.1%) families. Mutations were detected in the six genes: NPHS1 (2 out of 72), WT1 (2 out of 72), NPHS2, MYO1E, TRPC6, and INF2. Median age at onset was 4.1 years in patients without a mutation (range 0.5-18.8), and 3.2 years in those in whom the causative mutation was detected (range 0.1-14.3). Mutations in dominant genes presented with a median onset of 4.5 years (range 3.2-14.3). Mutations in recessive genes presented with a median onset of 0.5 years (range 0.1-3.2). CONCLUSION: Our molecular genetic diagnostic study identified underlying monogenic causes of steroid-resistant nephrotic syndrome in ~11% of patients with SRNS using a cost-effective technique. We delineated some of the therapeutic, diagnostic, and prognostic implications. Our study confirms that genetic testing is indicated in pediatric patients with SRNS.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Síndrome Nefrótico/congénito , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Síndrome Nefrótico/genética
16.
Am J Hum Genet ; 94(6): 884-90, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24814193

RESUMEN

Nephrotic syndrome (NS) is a genetically heterogeneous group of diseases that are divided into steroid-sensitive NS (SSNS) and steroid-resistant NS (SRNS). SRNS inevitably leads to end-stage kidney disease, and no curative treatment is available. To date, mutations in more than 24 genes have been described in Mendelian forms of SRNS; however, no Mendelian form of SSNS has been described. To identify a genetic form of SSNS, we performed homozygosity mapping, whole-exome sequencing, and multiplex PCR followed by next-generation sequencing. We thereby detected biallelic mutations in EMP2 (epithelial membrane protein 2) in four individuals from three unrelated families affected by SRNS or SSNS. We showed that EMP2 exclusively localized to glomeruli in the kidney. Knockdown of emp2 in zebrafish resulted in pericardial effusion, supporting the pathogenic role of mutated EMP2 in human NS. At the cellular level, we showed that knockdown of EMP2 in podocytes and endothelial cells resulted in an increased amount of CAVEOLIN-1 and decreased cell proliferation. Our data therefore identify EMP2 mutations as causing a recessive Mendelian form of SSNS.


Asunto(s)
Glicoproteínas de Membrana/genética , Mutación , Síndrome Nefrótico/genética , Alelos , Animales , Caveolina 1/metabolismo , Proliferación Celular , Preescolar , Mapeo Cromosómico , Células Endoteliales/patología , Regulación de la Expresión Génica , Sitios Genéticos , Homocigoto , Humanos , Lactante , Riñón/patología , Fallo Renal Crónico/etiología , Fallo Renal Crónico/genética , Glicoproteínas de Membrana/metabolismo , Síndrome Nefrótico/complicaciones , Pez Cebra/embriología , Pez Cebra/genética
17.
Nephrol Dial Transplant ; 31(11): 1802-1813, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26507970

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) represents the second most frequent cause of chronic kidney disease in the first three decades of life. It manifests histologically as focal segmental glomerulosclerosis (FSGS) and carries a 33% risk of relapse in a renal transplant. No efficient treatment exists. Identification of single-gene (monogenic) causes of SRNS has moved the glomerular epithelial cell (podocyte) to the center of its pathogenesis. Recently, mutations in >30 recessive or dominant genes were identified as causing monogenic forms of SRNS, thereby revealing the encoded proteins as essential for glomerular function. These findings helped define protein interaction complexes and functional pathways that could be targeted for treatment of SRNS. Very recently, it was discovered that in the surprisingly high fraction of ∼30% of all individuals who manifest with SRNS before 25 years of age, a causative mutation can be detected in one of the ∼30 different SRNS-causing genes. These findings revealed that SRNS and FSGS are not single disease entities but rather are part of a spectrum of distinct diseases with an identifiable genetic etiology. Mutation analysis should be offered to all individuals who manifest with SRNS before the age of 25 years, because (i) it will provide the patient and families with an unequivocal cause-based diagnosis, (ii) it may uncover a form of SRNS that is amenable to treatment (e.g. coenzyme Q10), (iii) it may allow avoidance of a renal biopsy procedure, (iv) it will further unravel the puzzle of pathogenic pathways of SRNS and (v) it will permit personalized treatment options for SRNS, based on genetic causation in way of 'precision medicine'.


Asunto(s)
Marcadores Genéticos/genética , Pruebas Genéticas/métodos , Mutación/genética , Síndrome Nefrótico/congénito , Análisis Mutacional de ADN , Humanos , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética
18.
J Am Soc Nephrol ; 26(6): 1279-89, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25349199

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) is the second most frequent cause of ESRD in the first two decades of life. Effective treatment is lacking. First insights into disease mechanisms came from identification of single-gene causes of SRNS. However, the frequency of single-gene causation and its age distribution in large cohorts are unknown. We performed exon sequencing of NPHS2 and WT1 for 1783 unrelated, international families with SRNS. We then examined all patients by microfluidic multiplex PCR and next-generation sequencing for all 27 genes known to cause SRNS if mutated. We detected a single-gene cause in 29.5% (526 of 1783) of families with SRNS that manifested before 25 years of age. The fraction of families in whom a single-gene cause was identified inversely correlated with age of onset. Within clinically relevant age groups, the fraction of families with detection of the single-gene cause was as follows: onset in the first 3 months of life (69.4%), between 4 and 12 months old (49.7%), between 1 and 6 years old (25.3%), between 7 and 12 years old (17.8%), and between 13 and 18 years old (10.8%). For PLCE1, specific mutations correlated with age of onset. Notably, 1% of individuals carried mutations in genes that function within the coenzyme Q10 biosynthesis pathway, suggesting that SRNS may be treatable in these individuals. Our study results should facilitate molecular genetic diagnostics of SRNS, etiologic classification for therapeutic studies, generation of genotype-phenotype correlations, and the identification of individuals in whom a targeted treatment for SRNS may be available.


Asunto(s)
Predisposición Genética a la Enfermedad/epidemiología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Síndrome Nefrótico/congénito , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Estudios de Cohortes , Femenino , Genes del Tumor de Wilms , Estudios de Asociación Genética , Genotipo , Heterocigoto , Humanos , Incidencia , Lactante , Masculino , Persona de Mediana Edad , Mutación , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Síndrome Nefrótico/fisiopatología , Linaje , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Medición de Riesgo , Adulto Joven
19.
Nat Genet ; 38(12): 1397-405, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17086182

RESUMEN

Nephrotic syndrome, a malfunction of the kidney glomerular filter, leads to proteinuria, edema and, in steroid-resistant nephrotic syndrome, end-stage kidney disease. Using positional cloning, we identified mutations in the phospholipase C epsilon gene (PLCE1) as causing early-onset nephrotic syndrome with end-stage kidney disease. Kidney histology of affected individuals showed diffuse mesangial sclerosis (DMS). Using immunofluorescence, we found PLCepsilon1 expression in developing and mature glomerular podocytes and showed that DMS represents an arrest of normal glomerular development. We identified IQ motif-containing GTPase-activating protein 1 as a new interaction partner of PLCepsilon1. Two siblings with a missense mutation in an exon encoding the PLCepsilon1 catalytic domain showed histology characteristic of focal segmental glomerulosclerosis. Notably, two other affected individuals responded to therapy, making this the first report of a molecular cause of nephrotic syndrome that may resolve after therapy. These findings, together with the zebrafish model of human nephrotic syndrome generated by plce1 knockdown, open new inroads into pathophysiology and treatment mechanisms of nephrotic syndrome.


Asunto(s)
Mutación , Síndrome Nefrótico/enzimología , Síndrome Nefrótico/genética , Fosfolipasas de Tipo C/genética , Animales , Niño , Preescolar , Clonación Molecular , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Genes Recesivos , Homocigoto , Humanos , Lactante , Riñón/enzimología , Riñón/patología , Masculino , Modelos Genéticos , Mutación Missense , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología , Fosfoinositido Fosfolipasa C , Ratas , Eliminación de Secuencia , Pez Cebra/genética
20.
Kidney Int ; 85(4): 880-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24257694

RESUMEN

Rare single-gene disorders cause chronic disease. However, half of the 6000 recessive single gene causes of disease are still unknown. Because recessive disease genes can illuminate, at least in part, disease pathomechanism, their identification offers direct opportunities for improved clinical management and potentially treatment. Rare diseases comprise the majority of chronic kidney disease (CKD) in children but are notoriously difficult to diagnose. Whole-exome resequencing facilitates identification of recessive disease genes. However, its utility is impeded by the large number of genetic variants detected. We here overcome this limitation by combining homozygosity mapping with whole-exome resequencing in 10 sib pairs with a nephronophthisis-related ciliopathy, which represents the most frequent genetic cause of CKD in the first three decades of life. In 7 of 10 sibships with a histologic or ultrasonographic diagnosis of nephronophthisis-related ciliopathy, we detect the causative gene. In six sibships, we identify mutations of known nephronophthisis-related ciliopathy genes, while in two additional sibships we found mutations in the known CKD-causing genes SLC4A1 and AGXT as phenocopies of nephronophthisis-related ciliopathy. Thus, whole-exome resequencing establishes an efficient, noninvasive approach towards early detection and causation-based diagnosis of rare kidney diseases. This approach can be extended to other rare recessive disorders, thereby providing accurate diagnosis and facilitating the study of disease mechanisms.


Asunto(s)
Pruebas Genéticas/métodos , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/genética , Adolescente , Adulto , Análisis Mutacional de ADN , Diagnóstico Precoz , Exoma , Genes Recesivos , Humanos , Lactante , Masculino , Mutación , Fenotipo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA