Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Clin Dev Immunol ; 2012: 695131, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23227086

RESUMEN

Inherited and acquired dysregulation of the complement alternative pathway plays an important role in multiple renal diseases. In recent years, the identification of disease-causing mutations and genetic variants in complement regulatory proteins has contributed significantly to our knowledge of the pathogenesis of complement associated glomerulopathies. In these diseases defective complement control leading to the deposition of activated complement products plays a key role. Consequently, complement-related glomerulopathies characterized by glomerular complement component 3 (C3) deposition in the absence of local immunoglobulin deposits are now collectively described by the term "C3 glomerulopathies." Therapeutic strategies for reestablishing complement regulation by either complement blockade with the anti-C5 monoclonal antibody eculizumab or plasma substitution have been successful in several cases of C3 glomerulopathies. However, further elucidation of the underlying defects in the alternative complement pathway is awaited to develop pathogenesis-specific therapies.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Enfermedades Renales/inmunología , Activación de Complemento/inmunología , Humanos , Enfermedades Renales/genética , Enfermedades Renales/metabolismo
2.
J Clin Invest ; 121(5): 2013-24, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21540551

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of end-stage renal failure. Identification of single-gene causes of SRNS has generated some insights into its pathogenesis; however, additional genes and disease mechanisms remain obscure, and SRNS continues to be treatment refractory. Here we have identified 6 different mutations in coenzyme Q10 biosynthesis monooxygenase 6 (COQ6) in 13 individuals from 7 families by homozygosity mapping. Each mutation was linked to early-onset SRNS with sensorineural deafness. The deleterious effects of these human COQ6 mutations were validated by their lack of complementation in coq6-deficient yeast. Furthermore, knockdown of Coq6 in podocyte cell lines and coq6 in zebrafish embryos caused apoptosis that was partially reversed by coenzyme Q10 treatment. In rats, COQ6 was located within cell processes and the Golgi apparatus of renal glomerular podocytes and in stria vascularis cells of the inner ear, consistent with an oto-renal disease phenotype. These data suggest that coenzyme Q10-related forms of SRNS and hearing loss can be molecularly identified and potentially treated.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Mutación , Síndrome Nefrótico/genética , Ubiquinona/genética , Animales , Células COS , Niño , Preescolar , Chlorocebus aethiops , Células HeLa , Pérdida Auditiva Sensorineural/complicaciones , Homocigoto , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/genética , Glomérulos Renales/metabolismo , Laminina/genética , Proteínas de la Membrana/genética , Síndrome Nefrótico/complicaciones , Fenotipo , Podocitos/metabolismo , Ratas , Proteínas WT1/genética , Pez Cebra
3.
Clin J Am Soc Nephrol ; 5(9): 1655-62, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20595692

RESUMEN

BACKGROUND AND OBJECTIVES: The risk of developing Wilms tumor (WT) can be present or absent in patients with nephrotic syndrome (NS) caused by WT1 mutations. Here, the genotype/phenotype correlation regarding the outcome and risk for WT in 52 patients from 51 families with NS due to WT1 mutations is described. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This study followed 19 patients with mutations in intron 9 splice donor site (KTS mutations), 27 patients with missense mutations, 4 patients with nonsense mutations, 1 patient with a splice site mutation in intron 8, and 1 patient with a deletion. RESULTS: Twenty-four different WT1 mutations were detected. Sixteen of the 19 patients with KTS mutations were females. These patients had isolated NS if karyotype was 46,XX and Frasier syndrome if karyotype was 46,XY. Patients with KTS mutations presented at a significantly older age and with a slower progression toward chronic kidney disease (CKD) stage 5, compared with missense mutations. Patients with nonsense mutations presented initially with WT. Six patients with missense mutations developed WT after the diagnosis of NS (interval-range from NS onset to WT of 0.1 to 1.4 years). CONCLUSIONS: (1) KTS mutations cause isolated NS with absence of WT in 46,XX females. (2) KTS mutations cause Frasier syndrome with gonadoblastoma risk in 46,XY phenotypic females. (3) KTS mutations cause NS with a slower progression when compared with missense mutations. (4) Missense mutations can occur with and without WT. (5) WT1 analysis is important in young patients with NS for early detection and tumor prophylaxis.


Asunto(s)
Síndrome de Frasier/genética , Genes del Tumor de Wilms , Gonadoblastoma/genética , Fallo Renal Crónico/genética , Neoplasias Renales/genética , Mutación , Síndrome Nefrótico/genética , Tumor de Wilms/genética , Edad de Inicio , Niño , Preescolar , Codón sin Sentido , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Síndrome de Frasier/etnología , Síndrome de Frasier/mortalidad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Gonadoblastoma/etnología , Gonadoblastoma/mortalidad , Humanos , Lactante , Intrones , Israel , Estimación de Kaplan-Meier , Cariotipificación , Fallo Renal Crónico/etnología , Fallo Renal Crónico/mortalidad , Neoplasias Renales/etnología , Neoplasias Renales/mortalidad , Masculino , Mutación Missense , Síndrome Nefrótico/etnología , Síndrome Nefrótico/mortalidad , Linaje , Fenotipo , Medición de Riesgo , Factores de Riesgo , Eliminación de Secuencia , Factores de Tiempo , Turquía , Estados Unidos , Tumor de Wilms/etnología , Tumor de Wilms/mortalidad
4.
Nephrol Dial Transplant ; 25(9): 2970-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20172850

RESUMEN

BACKGROUND: Recessive mutations in the NPHS1 gene encoding nephrin account for approximately 40% of infants with congenital nephrotic syndrome (CNS). CNS is defined as steroid-resistant nephrotic syndrome (SRNS) within the first 90 days of life. Currently, more than 119 different mutations of NPHS1 have been published affecting most exons. METHODS: We here performed mutational analysis of NPHS1 in a worldwide cohort of 67 children from 62 different families with CNS. RESULTS: We found bi-allelic mutations in 36 of the 62 families (58%) confirming in a worldwide cohort that about one-half of CNS is caused by NPHS1 mutations. In 26 families, mutations were homozygous, and in 10, they were compound heterozygous. In an additional nine patients from eight families, only one heterozygous mutation was detected. We detected 37 different mutations. Nineteen of the 37 were novel mutations (approximately 51.4%), including 11 missense mutations, 4 splice-site mutations, 3 nonsense mutations and 1 small deletion. In an additional patient with later manifestation, we discovered two further novel mutations, including the first one affecting a glycosylation site of nephrin. CONCLUSIONS: Our data hereby expand the spectrum of known mutations by 17.6%. Surprisingly, out of the two siblings with the homozygous novel mutation L587R in NPHS1, only one developed nephrotic syndrome before the age of 90 days, while the other one did not manifest until the age of 2 years. Both siblings also unexpectedly experienced an episode of partial remission upon steroid treatment.


Asunto(s)
Proteínas de la Membrana/genética , Mutación/genética , Síndrome Nefrótico/congénito , Síndrome Nefrótico/genética , Estudios de Cohortes , Exones/genética , Familia , Femenino , Genotipo , Salud Global , Heterocigoto , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Síndrome Nefrótico/patología , Fenotipo , Pronóstico
5.
Pediatr Nephrol ; 25(2): 261-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19876656

RESUMEN

Previous studies in children with focal segmental glomerulosclerosis (FSGS) and nephrotic syndrome (NS) in the USA have revealed inter-ethnic differences in their clinical presentation and outcome. However, ethnicity was based on self-identification rather than on molecular genetic data. Here, we show that genetic heterogeneity exists in self-identified Hispanic (Spanish-American) patients with steroid-resistant nephrotic syndrome (SRNS), as patients may be either of Caucasian or Mesoamerican (Native-American) genetic background. Twenty-one self-identified Hispanic patients with SRNS from 18 families were initially evaluated for mutations in the NPHS2 and WT1 genes. All patients resided and were cared for in the USA. We performed a total genome search for linkage in all Hispanic patients using 250K single nucleotide polymorphism microarrays, comparing Caucasian with Mesoamerican allele frequencies to determine regions of homozygosity by descent and to establish the correct allele frequency for each family. We found that only ten families (56%) of the 18 self-identified Hispanic families are genetically of Mesoamerican descent, whereas the other eight families (44%) are of Caucasian descent. Due to the small number of families examined, we were unable to draw any conclusion on the prevalence of NPHS2 and WT1 in this ethnic group, but the data do suggest that self-identification of ethnicity in Hispanic-American patients is not an adequate basis for genetic studies, as this cohort may represent not only patients of Mesoamerican origin but also patients of Caucasian origin. Thus, one needs to critically review previous studies of FSGS/SRNS patients that involved Hispanic patients as a group. Future larger studies may employ a total genome search for linkage to test self-identified Hispanic ethnicity for true Mesoamerican versus Caucasian ethnicity in order to generate valid genetic data.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Hispánicos o Latinos/genética , Síndrome Nefrótico/etnología , Síndrome Nefrótico/genética , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Bases de Datos Genéticas , Resistencia a Medicamentos , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genómica , Glomeruloesclerosis Focal y Segmentaria/etnología , Glomeruloesclerosis Focal y Segmentaria/patología , Hispánicos o Latinos/etnología , Humanos , Indígenas Sudamericanos/etnología , Indígenas Sudamericanos/genética , Lactante , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana , Síndrome Nefrótico/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Esteroides/uso terapéutico , Proteínas WT1/genética , Población Blanca/etnología , Población Blanca/genética
6.
PLoS One ; 4(11): e7771, 2009 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-19936226

RESUMEN

BACKGROUND: TRPC6, encoding a member of the transient receptor potential (TRP) superfamily of ion channels, is a calcium-permeable cation channel, which mediates capacitive calcium entry into the cell. Until today, seven different mutations in TRPC6 have been identified as a cause of autosomal-dominant focal segmental glomerulosclerosis (FSGS) in adults. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a novel TRPC6 mutation that leads to early onset FSGS. We identified one family in whom disease segregated with a novel TRPC6 mutation (M132T), that also affected pediatric individuals as early as nine years of age. Twenty-one pedigrees compatible with an autosomal-dominant mode of inheritance and biopsy-proven FSGS were selected from a worldwide cohort of 550 families with steroid resistant nephrotic syndrome (SRNS). Whole cell current recordings of the mutant TRPC6 channel, compared to the wild-type channel, showed a 3 to 5-fold increase in the average out- and inward TRPC6 current amplitude. The mean inward calcium current of M132T was 10-fold larger than that of wild-type TRPC6. Interestingly, M132T mutants also lacked time-dependent inactivation. Generation of a novel double mutant M132T/N143S did not further augment TRPC6 channel activity. CONCLUSIONS: In summary, our data shows that TRPC6 mediated FSGS can also be found in children. The large increase in channel currents and impaired channel inactivation caused by the M132T mutant leads to an aggressive phenotype that underlines the importance of calcium dose channeled through TRPC6.


Asunto(s)
Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/genética , Mutación , Canales Catiónicos TRPC/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Salud de la Familia , Femenino , Genes Dominantes , Humanos , Lactante , Masculino , Persona de Mediana Edad , Canal Catiónico TRPC6
7.
Kidney Int ; 75(7): 669-71, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19282856

RESUMEN

In steroid-resistant nephrotic syndrome (SRNS) Machuca et al. report that mutations of the recessive podocin gene cause adult-onset SRNS if the R229Q genetic variant occurs in a compound heterozygous state with another podocin mutation. Learning to tell apart the specific allele combinations of podocin mutations will be important for prognosis, genetic counseling in living related kidney donation, accurate etiologic classification within treatment studies, and the understanding of podocin function.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutación/fisiología , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Adulto , Edad de Inicio , Resistencia a Medicamentos , Humanos , Síndrome Nefrótico/diagnóstico , Pronóstico
8.
Pediatr Nephrol ; 24(7): 1399-401, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19205749

RESUMEN

Wilms' tumour suppressor gene 1 (WT1) encodes a transcription factor required for normal development of the genitourinary system. In the kidney, mutations in WT1 can cause diffuse mesangial sclerosis or focal segmental glomerulosclerosis. Here, we report on a girl with a mutation in WT1, who developed membranoproliferative glomerulonephritis (MPGN) 3 years after completion of treatment for Wilms' tumour. This finding extends the spectrum of glomerular disease seen with WT1 mutations and could have implications for the screening of children with MPGN.


Asunto(s)
Genes del Tumor de Wilms , Glomerulonefritis Membranoproliferativa/genética , Antineoplásicos/uso terapéutico , Niño , Preescolar , Terapia Combinada , Femenino , Glomerulonefritis Membranoproliferativa/complicaciones , Glomerulonefritis Membranoproliferativa/fisiopatología , Humanos , Inmunosupresores/uso terapéutico , Lactante , Trasplante de Riñón , Mutación , Nefrectomía , Vincristina/uso terapéutico , Tumor de Wilms/complicaciones , Tumor de Wilms/genética , Tumor de Wilms/cirugía
9.
PLoS Genet ; 5(1): e1000353, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19165332

RESUMEN

The identification of recessive disease-causing genes by homozygosity mapping is often restricted by lack of suitable consanguineous families. To overcome these limitations, we apply homozygosity mapping to single affected individuals from outbred populations. In 72 individuals of 54 kindred ascertained worldwide with known homozygous mutations in 13 different recessive disease genes, we performed total genome homozygosity mapping using 250,000 SNP arrays. Likelihood ratio Z-scores (ZLR) were plotted across the genome to detect ZLR peaks that reflect segments of homozygosity by descent, which may harbor the mutated gene. In 93% of cases, the causative gene was positioned within a consistent ZLR peak of homozygosity. The number of peaks reflected the degree of inbreeding. We demonstrate that disease-causing homozygous mutations can be detected in single cases from outbred populations within a single ZLR peak of homozygosity as short as 2 Mb, containing an average of only 16 candidate genes. As many specialty clinics have access to cohorts of individuals from outbred populations, and as our approach will result in smaller genetic candidate regions, the new strategy of homozygosity mapping in single outbred individuals will strongly accelerate the discovery of novel recessive disease genes.


Asunto(s)
Genes Recesivos , Análisis Mutacional de ADN , Reacciones Falso Positivas , Salud de la Familia , Femenino , Marcadores Genéticos , Genética de Población , Homocigoto , Humanos , Enfermedades Renales Quísticas/genética , Masculino , Modelos Genéticos , Síndrome Nefrótico/genética , Linaje , Esteroides/farmacología
10.
Pediatr Nephrol ; 24(5): 1065-70, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18853198

RESUMEN

Nephrotic syndrome presenting in the first year of life is often challenging, with substantial risk of progression to end-stage renal disease (ESRD). Focal segmental glomerulosclerosis (FSGS) comprises up to 20% of biopsy-proven glomerular disease in children and adults. We report on a 9-month-old infant who presented with nephrotic syndrome, hypertension and progressive deterioration of renal function due to FSGS. As immunosuppressive agents are often unsuccessful in treating this condition, we adopted renoprotection as the mainstay treatment for this patient, through rigorous control of blood pressure and proteinuria with a multi-drug regimen including renin-angiotensin axis blockade. Initially, there was partial improvement, with a gradual decline in proteinuria and a concomitant rise in the glomerular filtration rate, before the disease eventually passed into complete clinical and laboratory remission. We speculate that infants with steroid-resistant nephrotic syndrome due to FSGS may benefit from tight control of hypertension, mainly though early blockade of the renin-angiotensin axis. We believe that its renoprotecive mechanism counteracts the deleterious effects of both hypertension and proteinuria, thereby not only preventing progressive renal disease, but even paving the way for a remission, as in our patient. To the best of our knowledge, this is the first report of an infant with nephrotic syndrome (NS) due to FSGS that passed into complete remission while the patient was on renoprotective measures including the use of angiotensin-converting enzyme inhibitors (ACEis).


Asunto(s)
Angiotensinas/antagonistas & inhibidores , Antihipertensivos/uso terapéutico , Captopril/uso terapéutico , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Síndrome Nefrótico/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Albúminas/uso terapéutico , Amlodipino/uso terapéutico , Resistencia a Medicamentos/efectos de los fármacos , Quimioterapia Combinada , Furosemida/uso terapéutico , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Lactante , Labetalol/uso terapéutico , Masculino , Síndrome Nefrótico/etiología , Síndrome Nefrótico/patología , Prednisolona/farmacología , Inducción de Remisión , Sistema Renina-Angiotensina/fisiología
11.
Pediatr Nephrol ; 23(9): 1455-60, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18543005

RESUMEN

In African American (AA) children, focal segmental glomerulosclerosis (FSGS) is the leading cause of nephrotic syndrome (NS). It has been shown that AA children suffer from FSGS and steroid-resistant nephrotic syndrome (SRNS) at a higher frequency and with a more severe renal outcome in comparison with Caucasian children. Previous mutation analysis of large cohorts revealed that a high percentage of childhood SRNS is monogenic and that mutations in podocin (NPHS2) and Wilms' tumor gene 1 (WT1) account for approximately 30% of SRNS in children. To test whether AA children with SRNS have a similar or a higher mutation rate, we performed mutation analysis of NPHS2 and WT1 in a cohort of AA children with SRNS. Direct sequencing was carried out for all exons of NPHS2 and for exons 8 and 9 of WT1. We ascertained 18 children of AA descent in whom renal biopsy findings showed FSGS in 13 patients (72%) and minimal-change disease in five patients (28%). In both NPHS2 and WT1, no disease-causing mutations were detected. Our data strongly suggest that in AA children with SRNS, the frequency of NPHS2 mutations is much lower than in large cohorts of pediatric SRNS patients in the general population. Knowledge of mutation rate of NPHS2 in different populations of SRNS patients facilitates the physician in planning a suitable genetic screening strategy for patients.


Asunto(s)
Negro o Afroamericano/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutación , Síndrome Nefrótico/genética , Adolescente , Corticoesteroides/uso terapéutico , Niño , Preescolar , Resistencia a Medicamentos , Femenino , Genes del Tumor de Wilms , Humanos , Lactante , Recién Nacido , Riñón/patología , Masculino , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología
12.
Nephrol Dial Transplant ; 23(11): 3527-33, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18503012

RESUMEN

BACKGROUND: Congenital nephrotic syndrome (CNS) is de- fined as nephrotic syndrome that manifests at birth or within the first 3 months of life. Most patients develop end-stage renal disease (ESRD) within 2 to 3 years of life. CNS of the Finnish-type (CNF) features a rather specific renal histology and is caused by recessive mutations in the NPHS1 gene encoding nephrin, a major structural protein of the glomerular slit-diaphragm. So far, more than 80 different mutations of NPHS1 causing CNF have been published. METHODS: Here, we performed mutation analysis of NPHS1 by exon sequencing in a worldwide cohort of 32 children with CNS from 29 different families. RESULTS: Sixteen of the 29 families (55%) were found to have two disease-causing alleles in NPHS1. Two additional patients had a single heterozygous mutation in NPHS1. Thirteen of a total of 20 different mutations detected were novel (65%). These were five missense mutations, one nonsense mutation, three deletions, one insertion and three splice-site mutations. CONCLUSION: Our data expand the spectrum of known NPHS1 mutations by >15% in a worldwide cohort. Surprisingly, two patients with disease-causing mutations showed a relatively mild phenotype, as one patient had a partial remission with steroid treatment and one patient had normal renal function 1 year after the onset of disease. The increased number of known mutations will facilitate future studies into genotype/phenotype correlations.


Asunto(s)
Proteínas de la Membrana/genética , Mutación/genética , Síndrome Nefrótico/congénito , Síndrome Nefrótico/genética , Codón sin Sentido/genética , Estudios de Cohortes , Femenino , Eliminación de Gen , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Mutagénesis Insercional/genética , Mutación Missense/genética , Fenotipo
13.
Nephrol Dial Transplant ; 23(4): 1291-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18065803

RESUMEN

BACKGROUND AND OBJECTIVES: Diffuse mesangial sclerosis (DMS) is a histologically distinct variant of nephrotic syndrome (NS) that is characterized by early onset and by progression to end-stage kidney disease (ESKD). Besides syndromic DMS, isolated (non-syndromic) DMS (IDMS) has been described. The etiology and pathogenesis of DMS is not understood. We recently identified by positional cloning recessive mutations in the gene PLCE1/NPHS3 as a novel cause of IDMS. We demonstrated a role of PLCE1 in glomerulogenesis. Mutations in two other genes WT1 and LAMB2 may also cause IDMS. We therefore determine in this study the relative frequency of mutations in PLCE1, WT1 or LAMB2 as the cause of IDMS in a worldwide cohort. METHODS: We identified 40 children from 35 families with IDMS from a worldwide cohort of 1368 children with NS. All the subjects were analyzed for mutations in all exons of PLCE1 by multiplex capillary heteroduplex analysis and direct sequencing, by direct sequencing of exons 8 and 9 of WT1, and all the exons of LAMB2. RESULTS: The median (range) age at onset of NS was 11 (1-72) months. We detected truncating mutations in PLCE1 in 10/35 (28.6%) families and WT1 mutations in 3/35 (8.5%) families. We found no mutations in LAMB2. CONCLUSIONS: PLCE1 mutation is the most common cause of IDMS in this cohort. We previously reported that one child with truncating mutation in PLCE1 responded to cyclosporine therapy. If this observation is confirmed in a larger study, mutations in PLCE1 may serve as a biomarker for selecting patients with IDMS who may benefit from treatment.


Asunto(s)
Mesangio Glomerular/patología , Mutación , Nefroesclerosis/genética , Fosfoinositido Fosfolipasa C/genética , Biopsia , Preescolar , ADN , Análisis Mutacional de ADN , Exones , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lactante , Laminina/genética , Laminina/metabolismo , Masculino , Nefroesclerosis/metabolismo , Nefroesclerosis/patología , Fosfoinositido Fosfolipasa C/metabolismo , Reacción en Cadena de la Polimerasa , Pronóstico , Índice de Severidad de la Enfermedad , Proteínas WT1/genética , Proteínas WT1/metabolismo
14.
Nephrol Dial Transplant ; 22(8): 2201-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17442739

RESUMEN

BACKGROUND: The course of idiopathic membranous nephropathy (iMN) is variable in untreated patients. Accurate prediction of renal outcome would allow optimal treatment decisions. We demonstrated that urinary beta2-microglobulin (beta2M) predicted prognosis in iMN with high sensitivity and specificity. It has been suggested that focal segmental glomerulosclerosis (FSGS) is a discriminative parameter with independent prognostic value. METHODS: We selected patients with iMN biopsied between 1988 and 2002. Biopsies were analysed for the presence of FSGS, interstitial fibrosis and vascular lesions. Serum creatinine, creatinine clearance, proteinuria and blood pressure were recorded at baseline. Outcome variables included remission of proteinuria, renal death (RD) defined as serum creatinine >135 micromol/l or increase of serum creatinine of >50%, or end-stage renal disease (ESRD). In a subgroup of patients, urinary beta2-microglobulin (beta2M) was measured. RESULTS: We included 53 patients (33M, 20F). Mean age was 51 years, serum creatinine 99 micromol/l, and proteinuria 7.0 g/10 mmol creatinine. FSGS was present in 22 patients. These patients were characterized by a higher serum creatinine at time of biopsy (P = 0.035), more severe interstitial fibrosis (P = 0.001) and higher stage of membranous nephropathy (P = 0.001). During follow-up 24 patients developed RD, almost equally distributed between patients with and without FSGS. Renal survival was numerically, but not significantly, lower in patients with FSGS. In Cox proportional hazard analysis, only serum creatinine at the time of biopsy was an independent predictor of RD or ESRD (P < 0.001). In patients with known urinary beta2M, there was no significant correlation with FSGS score (P = 0.174). CONCLUSION: FSGS is not an accurate prognostic marker in iMN. Histological scoring of FSGS is inferior to measurement of urinary proteins in predicting renal outcome in iMN.


Asunto(s)
Glomerulonefritis Membranosa/complicaciones , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Enfermedades Renales/complicaciones , Adulto , Anciano , Creatinina/orina , Femenino , Glomerulonefritis Membranosa/diagnóstico , Humanos , Riñón/patología , Enfermedades Renales/diagnóstico , Fallo Renal Crónico/diagnóstico , Masculino , Persona de Mediana Edad , Pronóstico , Proteinuria/metabolismo , Proteinuria/orina , Resultado del Tratamiento , Microglobulina beta-2/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...