Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Clin J Am Soc Nephrol ; 16(5): 790-799, 2021 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32690722

RESUMO

Autosomal dominant polycystic kidney disease is the most common monogenic cause of ESKD. Genetic studies from patients and animal models have informed disease pathobiology and strongly support a "threshold model" in which cyst formation is triggered by reduced functional polycystin dosage below a critical threshold within individual tubular epithelial cells due to (1) germline and somatic PKD1 and/or PKD2 mutations, (2) mutations of genes (e.g., SEC63, SEC61B, GANAB, PRKCSH, DNAJB11, ALG8, and ALG9) in the endoplasmic reticulum protein biosynthetic pathway, or (3) somatic mosaicism. Genetic testing has the potential to provide diagnostic and prognostic information in cystic kidney disease. However, mutation screening of PKD1 is challenging due to its large size and complexity, making it both costly and labor intensive. Moreover, conventional Sanger sequencing-based genetic testing is currently limited in elucidating the causes of atypical polycystic kidney disease, such as within-family disease discordance, atypical kidney imaging patterns, and discordant disease severity between total kidney volume and rate of eGFR decline. In addition, environmental factors, genetic modifiers, and somatic mosaicism also contribute to disease variability, further limiting prognostication by mutation class in individual patients. Recent innovations in next-generation sequencing are poised to transform and extend molecular diagnostics at reasonable costs. By comprehensive screening of multiple cystic disease and modifier genes, targeted gene panel, whole-exome, or whole-genome sequencing is expected to improve both diagnostic and prognostic accuracy to advance personalized medicine in autosomal dominant polycystic kidney disease.


Assuntos
Rim Policístico Autossômico Dominante/genética , Animais , Testes Genéticos , Humanos , Mutação , Rim Policístico Autossômico Dominante/diagnóstico
2.
Clin J Am Soc Nephrol ; 16(7): 1101-1109, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33849932

RESUMO

The glomerular basement membrane is a vital component of the filtration barrier of the kidney and is primarily composed of a highly structured matrix of type IV collagen. Specific isoforms of type IV collagen, the α3(IV), α4(IV), and α5(IV) isoforms, assemble into trimers that are required for normal glomerular basement membrane function. Disruption or alteration in these isoforms leads to breakdown of the glomerular basement membrane structure and function and can lead to progressive CKD known as Alport syndrome. However, there is wide variability in phenotype among patients with mutations affecting type IV collagen that depends on a complex interplay of sex, genotype, and X-chromosome inactivation. This article reviews the genetic basis of collagen disorders of the kidney as well as potential treatments for these conditions, including direct alteration of the DNA, RNA therapies, and manipulation of collagen proteins.


Assuntos
Colágeno Tipo IV/genética , Nefrite Hereditária/genética , Nefrite Hereditária/terapia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Edição de Genes , Terapia Genética , Humanos , Chaperonas Moleculares/uso terapêutico , Nefrite Hereditária/complicações , Isoformas de Proteínas/genética , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Vacinas de mRNA/uso terapêutico
3.
Clin J Am Soc Nephrol ; 16(3): 458-466, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-32680915

RESUMO

Over the past decade, genome-wide association studies (GWAS) have emerged as a powerful tool to understand the genetic basis of complex traits in humans. The GWAS approach has been successfully applied to primary glomerular disorders, providing numerous novel insights into the genetic architecture of IgA nephropathy, membranous nephropathy, and steroid-sensitive nephrotic syndrome. IgA nephropathy appears to have a highly complex polygenic architecture, with nearly 20 genome-wide significant loci of small-to-moderate effects discovered to date. In contrast, the genetic susceptibility to membranous nephropathy and steroid-sensitive nephrotic syndrome appears to be driven by a small number of large-effect loci. The MHC locus on chromosome 6p21 is strongly associated with genetic susceptibility to all major types of immune-mediated glomerulopathies. However, a distinct set of classical HLA alleles is associated with each individual disease type, pinpointing to specific immune mechanisms underlying each of these conditions. Additional insights from the discovery of non-HLA risk loci reinforced the role of innate and adaptive immunity in the pathogenesis of these disorders, and highlighted important susceptibility overlaps between glomerular and other autoimmune and inflammatory conditions. Despite these initial successes, much larger GWAS and sequencing studies are still needed for each individual glomerular disease type. Increased power will be critical to comprehensively test for genetic effects across the full spectrum of allelic frequencies, to detect gene-gene and gene-environment interactions, and to potentially improve the performance of polygenic risk predictors. Moreover, the existing studies are limited mostly to European and East Asian populations, stressing the urgency to expand genetic discovery efforts to more diverse populations worldwide.


Assuntos
Estudo de Associação Genômica Ampla , Glomerulonefrite por IGA/genética , Glomerulonefrite Membranosa/genética , Síndrome Nefrótica/genética , Humanos , Síndrome Nefrótica/tratamento farmacológico , Esteroides/uso terapêutico
4.
Clin J Am Soc Nephrol ; 16(2): 294-303, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-32616495

RESUMO

Rates of many types of severe kidney disease are much higher in Black individuals than most other ethnic groups. Much of this disparity can now be attributed to genetic variants in the apoL1 (APOL1) gene found only in individuals with recent African ancestry. These variants greatly increase rates of hypertension-associated ESKD, FSGS, HIV-associated nephropathy, and other forms of nondiabetic kidney disease. We discuss the population genetics of APOL1 risk variants and the clinical spectrum of APOL1 nephropathy. We then consider clinical issues that arise for the practicing nephrologist caring for the patient who may have APOL1 kidney disease.


Assuntos
Apolipoproteína L1/genética , População Negra/genética , Nefropatias/etnologia , Nefropatias/genética , Nefropatia Associada a AIDS/genética , África/epidemiologia , Alelos , Nefropatias Diabéticas/genética , Variação Genética , Glomerulosclerose Segmentar e Focal/genética , Humanos , Hipertensão/complicações , Nefropatias/terapia , Falência Renal Crônica/genética , Transplante de Rim , Fatores de Risco
5.
Clin J Am Soc Nephrol ; 15(12): 1818-1828, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-32205319

RESUMO

The glomerular filtration barrier is a highly specialized capillary wall comprising fenestrated endothelial cells, podocytes, and an intervening basement membrane. In glomerular disease, this barrier loses functional integrity, allowing the passage of macromolecules and cells, and there are associated changes in both cell morphology and the extracellular matrix. Over the past 3 decades, there has been a transformation in our understanding about glomerular disease, fueled by genetic discovery, and this is leading to exciting advances in our knowledge about glomerular biology and pathophysiology. In current clinical practice, a genetic diagnosis already has important implications for management, ranging from estimating the risk of disease recurrence post-transplant to the life-changing advances in the treatment of atypical hemolytic uremic syndrome. Improving our understanding about the mechanistic basis of glomerular disease is required for more effective and personalized therapy options. In this review, we describe genotype and phenotype correlations for genetic disorders of the glomerular filtration barrier, with a particular emphasis on how these gene defects cluster by both their ontology and patterns of glomerular pathology.


Assuntos
Variação Genética , Barreira de Filtração Glomerular/patologia , Glomerulonefrite/genética , Células Endoteliais/patologia , Predisposição Genética para Doença , Membrana Basal Glomerular/patologia , Barreira de Filtração Glomerular/fisiopatologia , Glomerulonefrite/sangue , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Humanos , Fenótipo , Podócitos/patologia , Medição de Risco , Fatores de Risco
6.
Clin J Am Soc Nephrol ; 15(8): 1181-1189, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32041801

RESUMO

Whether individual results of genetic research studies ought to be disclosed to study participants has been debated in recent decades. Previously, the prevailing expert view discouraged the return of individual research results to participants because of the potential lack of analytic validity, questionable clinical validity and medical actionability, and questions about whether it is the role of research to provide participants with their data. With additional knowledge of participant perspectives and shifting views about the benefits of research and respect for participants, current expert consensus is moving toward support of returning such results. Significant ethical controversies remain, and there are many practical questions left to address, including appropriate procedures for returning results and the potential burden to clinicians when patients seek guidance about the clinical implications of research results. In this review, we describe current views regarding the return of genetic research results, including controversies and practical challenges, and consider the application of these issues to research on apolipoprotein L1 (APOL1), a gene recently associated with health disparities in kidney disease. Although this case is unique, it illustrates the complexities involved in returning results and highlights remaining questions.


Assuntos
Apolipoproteína L1/genética , Aconselhamento Genético , Testes Genéticos , Variação Genética , Falência Renal Crônica/genética , Projetos de Pesquisa , Sujeitos da Pesquisa , Aconselhamento Genético/ética , Predisposição Genética para Doença , Testes Genéticos/ética , Humanos , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/terapia , Fenótipo , Valor Preditivo dos Testes , Prognóstico , Medição de Risco , Fatores de Risco
7.
Clin J Am Soc Nephrol ; 15(7): 1028-1040, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32513823

RESUMO

Genome-wide association studies (GWASs) have facilitated the unbiased discovery of hundreds of genomic loci associated with CKD and kidney function. The vast majority of disease-associated DNA variants are noncoding. Those that are causal in CKD pathogenesis likely modulate transcription of target genes in a cell type-specific manner. To gain novel biological insights into mechanisms driving the development of CKD, the causal variants (which are usually not the most significant variant reported in a GWAS), their target genes, and causal cell types need to be identified. This functional validation requires a large number of new data sets, complex bioinformatics analyses, and experimental cellular and in vivo studies. Here, we review the basic principles and some of the current approaches being leveraged to assign functional significance to a genotype-phenotype association.


Assuntos
Biologia Computacional/métodos , Estudo de Associação Genômica Ampla , Mutação , Insuficiência Renal Crônica/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Epigênese Genética , Frequência do Gene , Genótipo , Humanos , Desequilíbrio de Ligação , Proteínas dos Microfilamentos/genética , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas
8.
Clin J Am Soc Nephrol ; 15(5): 695-703, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31992571

RESUMO

The kidney is a complex organ responsible for maintaining multiple aspects of homeostasis in the human body. The combination of distinct, yet interrelated, molecular functions across different cell types make the delineation of factors associated with loss or decline in kidney function challenging. Consequently, there has been a paucity of new diagnostic markers and treatment options becoming available to clinicians and patients in managing kidney diseases. A systems biology approach to understanding the kidney leverages recent advances in computational technology and methods to integrate diverse sets of data. It has the potential to unravel the interplay of multiple genes, proteins, and molecular mechanisms that drive key functions in kidney health and disease. The emergence of large, detailed, multilevel biologic and clinical data from national databases, cohort studies, and trials now provide the critical pieces needed for meaningful application of systems biology approaches in nephrology. The purpose of this review is to provide an overview of the current state in the evolution of the field. Recent successes of systems biology to identify targeted therapies linked to mechanistic biomarkers in the kidney are described to emphasize the relevance to clinical care and the outlook for improving outcomes for patients with kidney diseases.


Assuntos
Nefropatias , Nefrologia , Biologia de Sistemas , Animais , Biomarcadores/metabolismo , Predisposição Genética para Doença , Genômica , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/fisiopatologia , Nefropatias/terapia , Terapia de Alvo Molecular , Fenótipo , Medicina de Precisão , Prognóstico , Integração de Sistemas
9.
Clin J Am Soc Nephrol ; 15(2): 268-275, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31992573

RESUMO

The kidney is subject to a wide range of abnormalities, many of which have a significant hereditable component. Next generation sequencing is increasingly bringing the genetic drivers of Mendelian disease into focus at the base pair level, whereas inexpensive genotyping arrays have surveyed hundreds of thousands of individuals to identify common variants that predispose to kidney dysfunction. In this first article in a CJASN series on kidney genomics, we review how both rare and common variants contribute to kidney disease, explore how evolution may influence the genetic variants that affect kidney function, consider how genetic information is and will be used in the clinic, and identify some of the most important future directions for kidney disease research. Forthcoming articles in the series will elaborate on many of these themes.


Assuntos
Variação Genética , Nefropatias/genética , Rim/anormalidades , Animais , Evolução Molecular , Predisposição Genética para Doença , Genômica , Hereditariedade , Humanos , Rim/fisiopatologia , Nefropatias/diagnóstico , Nefropatias/fisiopatologia , Técnicas de Diagnóstico Molecular , Linhagem , Fenótipo , Fatores de Risco
10.
Clin J Am Soc Nephrol ; 15(4): 550-556, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-31992574

RESUMO

Methods to differentiate human pluripotent stem cells into kidney organoids were first introduced about 5 years ago, and since that time, the field has grown substantially. Protocols are producing increasingly complex three-dimensional structures, have been used to model human kidney disease, and have been adapted for high-throughput screening. Over this same time frame, technologies for massively parallel, single-cell RNA sequencing (scRNA-seq) have matured. Now, both of these powerful approaches are being combined to better understand how kidney organoids can be applied to the understanding of kidney development and disease. There are several reasons why this is a synergistic combination. Kidney organoids are complicated and contain many different cell types of variable maturity. scRNA-seq is an unbiased technology that can comprehensively categorize cell types, making it ideally suited to catalog all cell types present in organoids. These same characteristics also make scRNA-seq a powerful approach for quantitative comparisons between protocols, batches, and pluripotent cell lines as it becomes clear that reproducibility and quality can vary across all three variables. Lineage trajectories can be reconstructed using scRNA-seq data, enabling the rational adjustment of differentiation strategies to promote maturation of desired kidney cell types or inhibit differentiation of undesired off-target cell types. Here, we review the ways that scRNA-seq has been successfully applied in the organoid field and predict future applications for this powerful technique. We also review other developing single-cell technologies and discuss how they may be combined, using "multiomic" approaches, to improve our understanding of kidney organoid differentiation and usefulness in modeling development, disease, and toxicity testing.


Assuntos
Diferenciação Celular , Linhagem da Célula , Perfilação da Expressão Gênica , Rim/citologia , Células-Tronco Pluripotentes/fisiologia , RNA-Seq , Análise de Célula Única , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Rim/metabolismo , Organoides , Células-Tronco Pluripotentes/metabolismo , Transcriptoma
11.
Clin J Am Soc Nephrol ; 15(3): 404-411, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31636087

RESUMO

In this review of the application of proteomics and metabolomics to kidney disease research, we review key concepts, highlight illustrative examples, and outline future directions. The proteome and metabolome reflect the influence of environmental exposures in addition to genetic coding. Circulating levels of proteins and metabolites are dynamic and modifiable, and thus amenable to therapeutic targeting. Design and analytic considerations in proteomics and metabolomics studies should be tailored to the investigator's goals. For the identification of clinical biomarkers, adjustment for all potential confounding variables, particularly GFR, and strict significance thresholds are warranted. However, this approach has the potential to obscure biologic signals and can be overly conservative given the high degree of intercorrelation within the proteome and metabolome. Mass spectrometry, often coupled to up-front chromatographic separation techniques, is a major workhorse in both proteomics and metabolomics. High-throughput antibody- and aptamer-based proteomic platforms have emerged as additional, powerful approaches to assay the proteome. As the breadth of coverage for these methodologies continues to expand, machine learning tools and pathway analyses can help select the molecules of greatest interest and categorize them in distinct biologic themes. Studies to date have already made a substantial effect, for example elucidating target antigens in membranous nephropathy, identifying a signature of urinary peptides that adds prognostic information to urinary albumin in CKD, implicating circulating inflammatory proteins as potential mediators of diabetic nephropathy, demonstrating the key role of the microbiome in the uremic milieu, and highlighting kidney bioenergetics as a modifiable factor in AKI. Additional studies are required to replicate and expand on these findings in independent cohorts. Further, more work is needed to understand the longitudinal trajectory of select protein and metabolite markers, perform transomics analyses within merged datasets, and incorporate more kidney tissue-based investigation.


Assuntos
Nefropatias/metabolismo , Rim/metabolismo , Metaboloma , Metabolômica , Proteoma , Proteômica , Biomarcadores/metabolismo , Humanos , Nefropatias/diagnóstico , Nefropatias/etiologia , Nefropatias/terapia , Valor Preditivo dos Testes , Prognóstico , Medição de Risco , Fatores de Risco
12.
Clin J Am Soc Nephrol ; 15(11): 1643-1656, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-32409295

RESUMO

The past few years have seen major advances in genome-wide association studies (GWAS) of CKD and kidney function-related traits in several areas: increases in sample size from >100,000 to >1 million, enabling the discovery of >250 associated genetic loci that are highly reproducible; the inclusion of participants not only of European but also of non-European ancestries; and the use of advanced computational methods to integrate additional genomic and other unbiased, high-dimensional data to characterize the underlying genetic architecture and prioritize potentially causal genes and variants. Together with other large-scale biobank and genetic association studies of complex traits, these GWAS of kidney function-related traits have also provided novel insight into the relationship of kidney function to other diseases with respect to their genetic associations, genetic correlation, and directional relationships. A number of studies also included functional experiments using model organisms or cell lines to validate prioritized potentially causal genes and/or variants. In this review article, we will summarize these recent GWAS of CKD and kidney function-related traits, explain approaches for downstream characterization of associated genetic loci and the value of such computational follow-up analyses, and discuss related challenges along with potential solutions to ultimately enable improved treatment and prevention of kidney diseases through genetics.


Assuntos
Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/fisiopatologia , Albuminúria/urina , Animais , Creatinina/urina , Variação Genética , Estudo de Associação Genômica Ampla , Taxa de Filtração Glomerular , Humanos , Fenótipo
13.
Clin J Am Soc Nephrol ; 15(10): 1497-1510, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32646915

RESUMO

Expanded accessibility of genetic sequencing technologies, such as chromosomal microarray and massively parallel sequencing approaches, is changing the management of hereditary kidney diseases. Genetic causes account for a substantial proportion of pediatric kidney disease cases, and with increased utilization of diagnostic genetic testing in nephrology, they are now also detected at appreciable frequencies in adult populations. Establishing a molecular diagnosis can have many potential benefits for patient care, such as guiding treatment, familial testing, and providing deeper insights on the molecular pathogenesis of kidney diseases. Today, with wider clinical use of genetic testing as part of the diagnostic evaluation, nephrologists have the challenging task of selecting the most suitable genetic test for each patient, and then applying the results into the appropriate clinical contexts. This review is intended to familiarize nephrologists with the various technical, logistical, and ethical considerations accompanying the increasing utilization of genetic testing in nephrology care.


Assuntos
Testes Genéticos , Nefropatias/diagnóstico , Nefropatias/genética , Análise de Sequência de DNA , Adulto , Ensaios Clínicos como Assunto , Hibridização Genômica Comparativa , Exoma , Testes Genéticos/ética , Testes Genéticos/métodos , Genoma , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Nefropatias/terapia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo Genético
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA