Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Kidney Dis ; 79(3): 362-373, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34273436

RESUMO

RATIONALE & OBJECTIVE: Clinical trial data have demonstrated the efficacy of etelcalcetide for reducing parathyroid hormone (PTH) levels in hemodialysis (HD) patients. We provide a real-world summary of etelcalcetide utilization, dosing, effectiveness, and discontinuation since its US introduction in April 2017. STUDY DESIGN: New-user design within prospective cohort. SETTING & PARTICIPANTS: 2,596 new users of etelcalcetide from April 2017 through August 2019 in a national sample of adult maintenance HD patients in the US Dialysis Outcomes and Practice Patterns Study (DOPPS). PREDICTORS: Baseline PTH, prior cinacalcet use, initial etelcalcetide dose. OUTCOME: Trajectories of etelcalcetide dose, chronic kidney disease-mineral and bone disease (CKD-MBD) medications, and levels of PTH, serum calcium, and phosphorus in the 12 months after etelcalcetide initiation. ANALYTICAL APPROACH: Cumulative incidence methods for etelcalcetide discontinuation and linear generalized estimating equations for trajectory analyses. RESULTS: By August 2019, etelcalcetide prescriptions increased to 6% of HD patients from their first use in April 2017. Starting etelcalcetide dose was 15 mg/wk in 70% of patients and 7.5 mg/wk in 27% of patients; 49% of new users were prescribed cinacalcet in the prior 3 months. Etelcalcetide discontinuation was 9%, 17%, and 27% by 3, 6, and 12 months after initiation. One year after etelcalcetide initiation, mean PTH levels declined by 40%, from 948 to 566 pg/mL, and the proportion of patients with PTH within target (150-599 pg/mL) increased from 33% to 64% overall, from 0 to 60% among patients with baseline PTH ≥ 600 pg/mL, and from 30% to 63% among patients with prior cinacalcet use. The proportion of patients with serum phosphorus > 5.5 mg/dL decreased from 55% to 45%, while the prevalence of albumin-corrected serum calcium < 7.5 mg/dL remained at 1%-2%. There were increases in use of active vitamin D (from 77% to 87%) and calcium-based phosphate binders (from 41% to 50%) in the 12 months after etelcalcetide initiation. LIMITATIONS: Data are unavailable for provider dosing protocols, dose holds, or reasons for discontinuation. CONCLUSIONS: In the 12 months after etelcalcetide initiation, patients had large and sustained reductions in PTH levels. These results support the utility of etelcalcetide as an effective therapy to achieve the KDIGO-recommended guidelines for CKD-MBD markers in HD patients.


Assuntos
Doenças Ósseas , Distúrbio Mineral e Ósseo na Doença Renal Crônica , Hiperparatireoidismo Secundário , Insuficiência Renal Crônica , Adulto , Doenças Ósseas/complicações , Cálcio , Distúrbio Mineral e Ósseo na Doença Renal Crônica/tratamento farmacológico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Estudos de Coortes , Humanos , Hiperparatireoidismo Secundário/etiologia , Minerais , Hormônio Paratireóideo , Peptídeos , Estudos Prospectivos , Diálise Renal/efeitos adversos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia
2.
J Ren Nutr ; 31(1): 21-34, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32386937

RESUMO

Bone and mineral metabolism becomes dysregulated with progression of chronic kidney disease (CKD), and increasing levels of parathyroid hormone serve as an adaptive response to maintain normal phosphorus and calcium levels. In end-stage renal disease, this response becomes maladaptive and high levels of phosphorus may occur. We summarize strategies to control hyperphosphatemia based on a systematic literature review of clinical trial and real-world observational data on phosphorus control in hemodialysis patients with CKD-mineral bone disorder (CKD-MBD). These studies suggest that current management options (diet and lifestyle changes; regular dialysis treatment; and use of phosphate binders, vitamin D, calcimimetics) have their own benefits and limitations with variable clinical outcomes. A more integrated approach to phosphorus control in dialysis patients may be necessary, incorporating measurement of multiple biomarkers of CKD-MBD pathophysiology (calcium, phosphorus, and parathyroid hormone) and correlation between diet adjustments and CKD-MBD drugs, which may facilitate improved patient management.


Assuntos
Calcimiméticos/uso terapêutico , Quelantes/uso terapêutico , Dieta/métodos , Hiperfosfatemia/complicações , Hiperfosfatemia/terapia , Falência Renal Crônica/complicações , Vitamina D/uso terapêutico , Humanos
3.
Am J Physiol Renal Physiol ; 312(4): F577-F588, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28077374

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the fourth most common cause of end-stage renal disease. The disease course can be highly variable and treatment options are limited. To identify new therapeutic targets and prognostic biomarkers of disease, we conducted parallel discovery microarray profiling in normal and diseased human PKD1 cystic kidney cells. A total of 1,515 genes and 5 miRNA were differentially expressed by more than twofold in PKD1 cells. Functional enrichment analysis identified 30 dysregulated signaling pathways including the epidermal growth factor (EGF) receptor pathway. In this paper, we report that the EGF/ErbB family receptor ErbB4 is a major factor driving cyst growth in ADPKD. Expression of ErbB4 in vivo was increased in human ADPKD and Pkd1 cystic kidneys, both transcriptionally and posttranscriptionally by mir-193b-3p. Ligand-induced activation of ErbB4 drives cystic proliferation and expansion suggesting a pathogenic role in cystogenesis. Our results implicate ErbB4 activation as functionally relevant in ADPKD, both as a marker of disease activity and as a new therapeutic target in this major kidney disease.


Assuntos
Proliferação de Células , Perfilação da Expressão Gênica/métodos , Rim/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Rim Policístico Autossômico Dominante/genética , Receptor ErbB-4/genética , Animais , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Estudos de Associação Genética , Marcadores Genéticos , Predisposição Genética para Doença , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação , Neuregulina-1/farmacologia , Fenótipo , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Interferência de RNA , Receptor ErbB-4/agonistas , Receptor ErbB-4/metabolismo , Transdução de Sinais , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/genética , Ativação Transcricional , Transfecção , Regulação para Cima
4.
Hum Genomics ; 10(1): 37, 2016 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-27871310

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) causes progressive loss of renal function in adults as a consequence of the accumulation of cysts. ADPKD is the most common genetic cause of end-stage renal disease. Mutations in polycystin-1 occur in 87% of cases of ADPKD and mutations in polycystin-2 are found in 12% of ADPKD patients. The complexity of ADPKD has hampered efforts to identify the mechanisms underlying its pathogenesis. No current FDA (Federal Drug Administration)-approved therapies ameliorate ADPKD progression. RESULTS: We used the de Almeida laboratory's sensitive new transcriptogram method for whole-genome gene expression data analysis to analyze microarray data from cell lines developed from cell isolates of normal kidney and of both non-cystic nephrons and cysts from the kidney of a patient with ADPKD. We compared results obtained using standard Ingenuity Volcano plot analysis, Gene Set Enrichment Analysis (GSEA) and transcriptogram analysis. Transcriptogram analysis confirmed the findings of Ingenuity, GSEA, and published analysis of ADPKD kidney data and also identified multiple new expression changes in KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways related to cell growth, cell death, genetic information processing, nucleotide metabolism, signal transduction, immune response, response to stimulus, cellular processes, ion homeostasis and transport and cofactors, vitamins, amino acids, energy, carbohydrates, drugs, lipids, and glycans. Transcriptogram analysis also provides significance metrics which allow us to prioritize further study of these pathways. CONCLUSIONS: Transcriptogram analysis identifies novel pathways altered in ADPKD, providing new avenues to identify both ADPKD's mechanisms of pathogenesis and pharmaceutical targets to ameliorate the progression of the disease.


Assuntos
Rim Policístico Autossômico Dominante/metabolismo , Transcriptoma , Adulto , Estudos de Casos e Controles , Linhagem Celular , Perfilação da Expressão Gênica , Ontologia Genética , Humanos , Masculino , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
5.
Am J Kidney Dis ; 66(4): 564-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25960302

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common potentially life-threatening monogenic disorder in humans, characterized by progressive development and expansion of fluid-filled cysts in the kidneys and other organs. Ongoing cyst growth leads to progressive kidney enlargement, whereas kidney function remains stable for decades as a result of hyperfiltration and compensation by unaffected nephrons. Kidney function irreversibly declines only in the late stages of the disease, when most of the parenchyma is lost to cystic and fibrotic tissue and the remaining compensatory capacity is overwhelmed. Hence, conventional kidney function measures, such as glomerular filtration rate, do not adequately assess disease progression in ADPKD, especially in its early stages. Given the recent development of potential targeted therapies in ADPKD, it has become critically important to identify relevant biomarkers that can be used to determine the degree of disease progression and evaluate the effects of therapeutic interventions on the course of the disease. We review the current evidence to provide an informed perspective on whether total kidney volume (TKV) is a suitable biomarker for disease progression and whether TKV can be used as an efficacy end point in clinical trials. We conclude that because cystogenesis is the central factor leading to kidney enlargement, TKV appears to be an appropriate biomarker and is gaining wider acceptance. Several studies have identified TKV as a relevant imaging biomarker for monitoring and predicting disease progression and support its use as a prognostic end point in clinical trials.


Assuntos
Taxa de Filtração Glomerular/fisiologia , Rim Policístico Autossômico Dominante/patologia , Rim Policístico Autossômico Dominante/terapia , Biomarcadores/sangue , Progressão da Doença , Feminino , Humanos , Testes de Função Renal , Masculino , Tamanho do Órgão/fisiologia , Rim Policístico Autossômico Dominante/genética , Prognóstico , Medição de Risco , Índice de Gravidade de Doença
6.
J Am Soc Nephrol ; 25(11): 2399-418, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24925719

RESUMO

Autosomal dominant polycystic kidney disease is a genetic disorder associated with substantial variability in its natural course within and between affected families. Understanding predictors for rapid progression of this disease has become increasingly important with the emergence of potential new treatments. This systematic review of the literature since 1988 evaluates factors that may predict and/or effect autosomal dominant polycystic kidney disease progression. Predicting factors associated with early adverse structural and/or functional outcomes are considered. These factors include PKD1 mutation (particularly truncating mutation), men, early onset of hypertension, early and frequent gross hematuria, and among women, three or more pregnancies. Increases in total kidney volume and decreases in GFR and renal blood flow greater than expected for a given age also signify rapid disease progression. Concerning laboratory markers include overt proteinuria, macroalbuminuria, and perhaps, elevated serum copeptin levels in affected adults. These factors and others may help to identify patients with autosomal dominant polycystic kidney disease who are most likely to benefit from early intervention with novel treatments.


Assuntos
Rim/patologia , Rim/fisiopatologia , Rim Policístico Autossômico Dominante , Canais de Cátion TRPP/genética , Progressão da Doença , Humanos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Rim Policístico Autossômico Dominante/fisiopatologia , Valor Preditivo dos Testes
7.
Kidney Int ; 85(2): 383-92, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23760289

RESUMO

Mutations to PKD1 and PKD2 are associated with autosomal dominant polycystic kidney disease (ADPKD). The absence of apparent PKD1/PKD2 linkage in five published European or North American families with ADPKD suggested a third locus, designated PKD3. Here we re-evaluated these families by updating clinical information, re-sampling where possible, and mutation screening for PKD1/PKD2. In the French-Canadian family, we identified PKD1: p.D3782_V3783insD, with misdiagnoses in two individuals and sample contamination explaining the lack of linkage. In the Portuguese family, PKD1: p.G3818A segregated with the disease in 10 individuals in three generations with likely misdiagnosis in one individual, sample contamination, and use of distant microsatellite markers explaining the linkage discrepancy. The mutation PKD2: c.213delC was found in the Bulgarian family, with linkage failure attributed to false positive diagnoses in two individuals. An affected son, but not the mother, in the Italian family had the nonsense mutation PKD1: p.R4228X, which appeared de novo in the son, with simple cysts probably explaining the mother's phenotype. No likely mutation was found in the Spanish family, but the phenotype was atypical with kidney atrophy in one case. Thus, re-analysis does not support the existence of a PKD3 in ADPKD. False positive diagnoses by ultrasound in all resolved families shows the value of mutation screening, but not linkage, to understand families with discrepant data.


Assuntos
Loci Gênicos , Mutação , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Adolescente , Adulto , Idoso , Canadá , Criança , Análise Mutacional de DNA , Erros de Diagnóstico , Europa (Continente) , Reações Falso-Positivas , Feminino , Ligação Genética , Predisposição Genética para Doença , Testes Genéticos/métodos , Haplótipos , Hereditariedade , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Rim Policístico Autossômico Dominante/diagnóstico por imagem , Valor Preditivo dos Testes , Ultrassonografia , Adulto Jovem
8.
J Mol Cell Cardiol ; 58: 199-208, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23376035

RESUMO

Mutations in PKD1 and PKD2, the genes encoding the proteins polycystin-1 (PC1) and polycystin-2 (PC2), cause autosomal dominant polycystic kidney disease (ADPKD). Although the leading cause of mortality in ADPKD is cardiovascular disease, the relationship between these conditions remains poorly understood. PC2 is an intracellular calcium channel expressed in renal epithelial cells and in cardiomyocytes, and is thus hypothesized to modulate intracellular calcium signaling and affect cardiac function. Our first aim was to study cardiac function in a zebrafish model lacking PC2 (pkd2 mutants). Next, we aimed to explore the relevance of this zebrafish model to human ADPKD by examining the Mayo Clinic's ADPKD database for an association between ADPKD and idiopathic dilated cardiomyopathy (IDCM). Pkd2 mutant zebrafish showed low cardiac output and atrioventricular block. Isolated pkd2 mutant hearts displayed impaired intracellular calcium cycling and calcium alternans. These results indicate heart failure in the pkd2 mutants. In human ADPKD patients, we found IDCM to coexist frequently with ADPKD. This association was strongest in patients with PKD2 mutations. Our results demonstrate that PC2 modulates intracellular calcium cycling, contributing to the development of heart failure. In human subjects we found an association between ADPKD and IDCM and suggest that PKD mutations contribute to the development of heart failure.


Assuntos
Cardiomiopatia Dilatada/genética , Proteínas de Transporte/genética , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Proteínas de Peixe-Zebra/genética , Animais , Cálcio/metabolismo , Sinalização do Cálcio/genética , Cardiomiopatia Dilatada/fisiopatologia , Predisposição Genética para Doença , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Podócitos/metabolismo , Podócitos/patologia , Rim Policístico Autossômico Dominante/fisiopatologia , Canais de Cátion TRPP/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
9.
Hum Mol Genet ; 20(13): 2524-34, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21493627

RESUMO

Meckel syndrome (MKS) is an embryonic lethal, autosomal recessive disorder characterized by polycystic kidney disease, central nervous system defects, polydactyly and liver fibrosis. This disorder is thought to be associated with defects in primary cilia; therefore, it is classed as a ciliopathy. To date, six genes have been commonly associated with MKS (MKS1, TMEM67, TMEM216, CEP290, CC2D2A and RPGRIP1L). However, mutation screening of these genes revealed two mutated alleles in only just over half of our MKS cohort (46 families), suggesting an even greater level of genetic heterogeneity. To explore the full genetic complexity of MKS, we performed exon-enriched next-generation sequencing of 31 ciliopathy genes in 12 MKS pedigrees using RainDance microdroplet-PCR enrichment and IlluminaGAIIx next-generation sequencing. In family M456, we detected a splice-donor site change in a novel MKS gene, B9D1. The B9D1 protein is structurally similar to MKS1 and has been shown to be of importance for ciliogenesis in Caenorhabditis elegans. Reverse transcriptase-PCR analysis of fetal RNA revealed, hemizygously, a single smaller mRNA product with a frameshifting exclusion of B9D1 exon 4. ArrayCGH showed that the second mutation was a 1.713 Mb de novo deletion completely deleting the B9D1 allele. Immunofluorescence analysis highlighted a significantly lower level of ciliated patient cells compared to controls, confirming a role for B9D1 in ciliogenesis. The fetus inherited an additional likely pathogenic novel missense change to a second MKS gene, CEP290; p.R2210C, suggesting oligogenic inheritance in this disorder.


Assuntos
Transtornos da Motilidade Ciliar/genética , Encefalocele/genética , Éxons/genética , Doenças Renais Policísticas/genética , Proteínas/genética , Deleção de Sequência/genética , Sequência de Aminoácidos , Sequência de Bases , Cílios/genética , Cílios/patologia , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Proteínas do Citoesqueleto , Encefalocele/metabolismo , Encefalocele/patologia , Feminino , Feto , Fibroblastos/metabolismo , Ordem dos Genes , Humanos , Espaço Intracelular/metabolismo , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Linhagem , Fenótipo , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Polimorfismo de Nucleotídeo Único/genética , Transporte Proteico/genética , Retinose Pigmentar , Alinhamento de Sequência
10.
BMC Nephrol ; 14: 190, 2013 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-24011172

RESUMO

BACKGROUND: Polycystic kidney diseases (PKD) are a group of monogenic disorders that are inherited dominantly (autosomal dominant PKD; ADPKD) or recessively, including, autosomal recessive PKD (ARPKD). A number of recessive, syndromic disorders also involve PKD but have a range of pleiotropic phenotypes beyond the kidney, and are enriched in consanguineous families. CASE PRESENTATION: We describe here a consanguineous Iranian pedigree in which PKD was diagnosed in four generations, but also included cases with additional abnormalities, including mental retardation. We employed molecular screening to reveal the etiology of the PKD. Since the PKD seemed to be dominantly inherited, molecular diagnostics was performed by direct sequencing of the ADPKD genes, PKD1 and PKD2. Clinical and imaging data was collected on family members. The sequence analysis revealed a PKD2 single base-pair deletion, c.1142delG, and segregation was demonstrated in 16 PKD patients from different branches of the family. In keeping with other reports, the PKD2 phenotype in this family was overall mild, and characterized by conserved kidney function, although 12 cases had some evidence of renal insufficiency. Several younger mutation carriers had borderline or no clinical characteristics of ADPKD, while a patient that required a renal transplant at 14 y did not have the PKD2 mutation. CONCLUSIONS: The molecular analysis of an Iranian family showed that the PKD was due to a PKD2 mutation. The identification of the causative mutation allowed an accurate diagnosis in a number of individuals with equivocal imaging data. Consequently, these patients could be followed appropriately as at-risk individuals. In addition, the PKD2 diagnosis ruled out a syndromic form of PKD as the cause of the additional phenotypes in the family.


Assuntos
Marcadores Genéticos/genética , Predisposição Genética para Doença/genética , Doenças Renais Policísticas/diagnóstico , Doenças Renais Policísticas/genética , Polimorfismo de Nucleotídeo Único/genética , Canais de Cátion TRPP/genética , Adulto , Idoso , Consanguinidade , Feminino , Testes Genéticos/métodos , Humanos , Irã (Geográfico) , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular/métodos , Mutação/genética , Linhagem
11.
J Am Soc Nephrol ; 23(5): 915-33, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22383692

RESUMO

Mutations in two large multi-exon genes, PKD1 and PKD2, cause autosomal dominant polycystic kidney disease (ADPKD). The duplication of PKD1 exons 1-32 as six pseudogenes on chromosome 16, the high level of allelic heterogeneity, and the cost of Sanger sequencing complicate mutation analysis, which can aid diagnostics of ADPKD. We developed and validated a strategy to analyze both the PKD1 and PKD2 genes using next-generation sequencing by pooling long-range PCR amplicons and multiplexing bar-coded libraries. We used this approach to characterize a cohort of 230 patients with ADPKD. This process detected definitely and likely pathogenic variants in 115 (63%) of 183 patients with typical ADPKD. In addition, we identified atypical mutations, a gene conversion, and one missed mutation resulting from allele dropout, and we characterized the pattern of deep intronic variation for both genes. In summary, this strategy involving next-generation sequencing is a model for future genetic characterization of large ADPKD populations.


Assuntos
Mutação , Rim Policístico Autossômico Dominante/genética , Análise de Sequência de DNA/métodos , Canais de Cátion TRPP/genética , Processamento Eletrônico de Dados , Humanos , Reação em Cadeia da Polimerase
12.
Nat Genet ; 30(3): 259-69, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11919560

RESUMO

Autosomal recessive polycystic kidney disease (ARPKD) is characterized by dilation of collecting ducts and by biliary dysgenesis and is an important cause of renal- and liver-related morbidity and mortality. Genetic analysis of a rat with recessive polycystic kidney disease revealed an orthologous relationship between the rat locus and the ARPKD region in humans; a candidate gene was identified. A mutation was characterized in the rat and screening the 66 coding exons of the human ortholog (PKHD1) in 14 probands with ARPKD revealed 6 truncating and 12 missense mutations; 8 of the affected individuals were compound heterozygotes. The PKHD1 transcript, approximately 16 kb long, is expressed in adult and fetal kidney, liver and pancreas and is predicted to encode a large novel protein, fibrocystin, with multiple copies of a domain shared with plexins and transcription factors. Fibrocystin may be a receptor protein that acts in collecting-duct and biliary differentiation.


Assuntos
Mutação , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética , Adulto , Sequência de Aminoácidos , Animais , Clonagem Molecular , Feminino , Testes Genéticos , Heterozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Rim Policístico Autossômico Recessivo/diagnóstico , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/química , Homologia de Sequência de Aminoácidos
13.
Hum Genet ; 129(3): 345-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21274727

RESUMO

The autosomal recessive polycystic kidney disease (ARPKD) gene, PKHD1, has been implicated in the genesis or growth of colorectal adenocarcinoma, as a high level of somatic mutations was found in colorectal tumor tissue. To determine whether carriers of a single PKHD1 mutation are at increased risk of colorectal carcinoma, we assessed the prevalence of the commonest European mutation, T36M. First, we assayed a European cohort of ARPKD patients and found T36M was responsible for 13.1% of mutations. We then investigated two European cohorts with colorectal adenocarcinoma versus two control cohorts of similar age and gender. Screening for the most common PKHD1 mutation, T36M, we detected 15:3,603 (0.42%) controls versus 1:3,767 (0.027%) colorectal cancer individuals, indicating that heterozygous PKHD1 mutations are not a risk factor and are protective (p=0.0002). We also show that the carriage rate for PKHD1 mutations in the European population is higher than previous accepted at 3.2% (1:31 genomes).


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Mutação em Linhagem Germinativa , Receptores de Superfície Celular/genética , Adenocarcinoma/epidemiologia , Idoso , Estudos de Coortes , Neoplasias Colorretais/epidemiologia , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , População Branca/genética
14.
Mol Genet Metab ; 104(4): 457-69, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21917492

RESUMO

The detection of single nucleotide polymorphisms (SNPs) and insertion/deletions (indels) with precision from high-throughput data remains a significant bioinformatics challenge. Accurate detection is necessary before next-generation sequencing can routinely be used in the clinic. In research, scientific advances are inhibited by gaps in data, exemplified by the underrepresented discovery of rare variants, variants in non-coding regions and indels. The continued presence of false positives and false negatives prevents full automation and requires additional manual verification steps. Our methodology presents applications of both pattern recognition and sensitivity analysis to eliminate false positives and aid in the detection of SNP/indel loci and genotypes from high-throughput data. We chose FK506-binding protein 51(FKBP5) (6p21.31) for our clinical target because of its role in modulating pharmacological responses to physiological and synthetic glucocorticoids and because of the complexity of the genomic region. We detected genetic variation across a 160 kb region encompassing FKBP5. 613 SNPs and 57 indels, including a 3.3 kb deletion were discovered. We validated our method using three independent data sets and, with Sanger sequencing and Affymetrix and Illumina microarrays, achieved 99% concordance. Furthermore we were able to detect 267 novel rare variants and assess linkage disequilibrium. Our results showed both a sensitivity and specificity of 98%, indicating near perfect classification between true and false variants. The process is scalable and amenable to automation, with the downstream filters taking only 1.5h to analyze 96 individuals simultaneously. We provide examples of how our level of precision uncovered the interactions of multiple loci, their predicted influences on mRNA stability, perturbations of the hsp90 binding site, and individual variation in FKBP5 expression. Finally we show how our discovery of rare variants may change current conceptions of evolution at this locus.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Mutação INDEL , Reconhecimento Automatizado de Padrão , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a Tacrolimo/genética , Regiões 3' não Traduzidas , Algoritmos , Sequência de Bases , Simulação por Computador , Variação Genética , Haplótipos , Humanos , Desequilíbrio de Ligação , Modelos Genéticos , Conformação de Ácido Nucleico , Estabilidade de RNA , Elementos de Resposta , Sensibilidade e Especificidade , Análise de Sequência de DNA , População Branca
15.
Transfus Apher Sci ; 45(3): 281-5, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22019727

RESUMO

BACKGROUND: Large use of allogeneic red blood cell concentrates (RBCc), albeit necessary in major surgery, may influence patients' outcome. DESIGN AND METHODS: We introduced an integrated strategy including patients' evaluation and supplementation associated with autologous blood collection and saving to support major elective surgery at our hospital since 2008. After 2 years of stabilization of this approach, we analyzed the results obtained in 2010 in terms of allogeneic blood usage and reduction of transfusion of stored RBCc. RESULTS: Analyzing 2010 results we found that usage of total autologous RBCc units was increased by 2.2 folds, of "not stored" autologous RBCc units by 2.4 folds and of allogeneic RBCc unit transfusion reduced by 65%. The significant reduction in the number of transfused allogeneic RBCc units associated with the use of "fresher" blood could prevent patients' complications due to immunomodulation and biologic/metabolic disregulation.


Assuntos
Transfusão de Sangue Autóloga/métodos , Transfusão de Eritrócitos/métodos , Procedimentos Cirúrgicos Eletivos , Feminino , Humanos , Masculino , Estudos Retrospectivos
16.
J Am Soc Nephrol ; 21(6): 1052-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20431041

RESUMO

There are no proven, effective therapies for polycystic kidney disease (PKD) or polycystic liver disease (PLD). We enrolled 42 patients with severe PLD resulting from autosomal dominant PKD (ADPKD) or autosomal dominant PLD (ADPLD) in a randomized, double-blind, placebo-controlled trial of octreotide, a long-acting somatostatin analogue. We randomly assigned 42 patients in a 2:1 ratio to octreotide LAR depot (up to 40 mg every 28+/-5 days) or placebo for 1 year. The primary end point was percent change in liver volume from baseline to 1 year, measured by MRI. Secondary end points were changes in total kidney volume, GFR, quality of life, safety, vital signs, and clinical laboratory tests. Thirty-four patients had ADPKD, and eight had ADPLD. Liver volume decreased by 4.95%+/-6.77% in the octreotide group but remained practically unchanged (+0.92%+/-8.33%) in the placebo group (P=0.048). Among patients with ADPKD, total kidney volume remained practically unchanged (+0.25%+/-7.53%) in the octreotide group but increased by 8.61%+/-10.07% in the placebo group (P=0.045). Changes in GFR were similar in both groups. Octreotide was well tolerated; treated individuals reported an improved perception of bodily pain and physical activity. In summary, octreotide slowed the progressive increase in liver volume and total kidney volume, improved health perception among patients with PLD, and had an acceptable side effect profile.


Assuntos
Hepatopatias/tratamento farmacológico , Octreotida/uso terapêutico , Rim Policístico Autossômico Dominante/tratamento farmacológico , Somatostatina/análogos & derivados , Adulto , Idoso , Método Duplo-Cego , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Hepatopatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Octreotida/efeitos adversos , Octreotida/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Rim Policístico Autossômico Dominante/fisiopatologia , Resultado do Tratamento
18.
Hum Mutat ; 30(4): 493-5, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19306393

RESUMO

Several Locus-Specific DataBases (LSDBs) have recently been approached by larger, more general data repositories (including NCBI and UCSC) with the request to share the DNA variant data they have collected. Within the Human Genome Variation Society (HGVS) a document was generated summarizing the issues related to these requests. The document has been circulated in the HGVS/LSDB community and was discussed extensively. Here we summarize these discussions and present the concluded recommendations for LSDB data sharing with central repositories.


Assuntos
Bases de Dados Genéticas , Variação Genética , Genoma Humano/genética , Disseminação de Informação , Biologia Computacional/métodos , Biologia Computacional/normas , Humanos , Mutação , Polimorfismo de Nucleotídeo Único
19.
Kidney Int ; 75(8): 848-55, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19165178

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) caused by mutations in PKD1 is significantly more severe than PKD2. Typically, ADPKD presents in adulthood but is rarely diagnosed in utero with enlarged, echogenic kidneys. Somatic mutations are thought crucial for cyst development, but gene dosage is also important since animal models with hypomorphic alleles develop cysts, but are viable as homozygotes. We screened for mutations in PKD1 and PKD2 in two consanguineous families and found PKD1 missense variants predicted to be pathogenic. In one family, two siblings homozygous for R3277C developed end stage renal disease at ages 75 and 62 years, while six heterozygotes had few cysts. In the other family, the father and two children with moderate to severe disease were homozygous for N3188S. In both families homozygous disease was associated with small cysts of relatively uniform size while marked cyst heterogeneity is typical of ADPKD. In another family, one patient diagnosed in childhood was found to be a compound heterozygote for the PKD1 variants R3105W and R2765C. All three families had evidence of developmental defects of the collecting system. Three additional ADPKD families with in utero onset had a truncating mutation in trans with either R3277C or R2765C. These cases suggest the presence of incompletely penetrant PKD1 alleles. The alleles alone may result in mild cystic disease; two such alleles cause typical to severe disease; and, in combination with an inactivating allele, are associated with early onset disease. Our study indicates that the dosage of functional PKD1 protein may be critical for cyst initiation.


Assuntos
Alelos , Dosagem de Genes , Penetrância , Canais de Cátion TRPP/genética , Cistos/genética , Análise Mutacional de DNA , Saúde da Família , Genótipo , Humanos , Falência Renal Crônica , Mutação de Sentido Incorreto , Linhagem
20.
Nephrol Dial Transplant ; 24(10): 3089-96, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19458060

RESUMO

BACKGROUND: Mutations in the TRPC6 gene have been reported in six families with adult-onset (17-57 years) autosomal dominant focal segmental glomerulosclerosis (FSGS). Electrophysiology studies confirmed augmented calcium influx only in three of these six TRPC6 mutations. To date, the role of TRPC6 in childhood and adulthood non-familial forms is unknown. METHODS: TRPC6 mutation analysis was performed by direct sequencing in 130 Spanish patients from 115 unrelated families with FSGS. An in silico scoring matrix was developed to evaluate the pathogenicity of amino acid substitutions, by using the bio-physical and bio-chemical differences between wild-type and mutant amino acid, the evolutionary conservation of the amino acid residue in orthologues, homologues and defined domains, with the addition of contextual information. RESULTS: Three new missense substitutions were identified in two clinically non-familial cases and in one familial case. The analysis by means of this scoring system allowed us to classify these variants as likely pathogenic mutations. One of them was detected in a female patient with unusual clinical features: mesangial proliferative FSGS in childhood (7 years) and partial response to immunosupressive therapy (CsA + MMF). Asymptomatic carriers of this likely mutation were found within her family. CONCLUSIONS: We describe for the first time TRPC6 mutations in children and adults with non-familial FSGS. It seems that TRPC6 is a gene with a very variable penetrance that may contribute to glomerular diseases in a multi-hit setting.


Assuntos
Glomerulosclerose Segmentar e Focal/genética , Canais de Cátion TRPC/genética , Adolescente , Adulto , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Pessoa de Meia-Idade , Canal de Cátion TRPC6 , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA