Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
bioRxiv ; 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38915495

RESUMO

Inverted formin-2 (INF2) gene mutations are among the most common causes of genetic focal segmental glomerulosclerosis (FSGS) with or without Charcot-Marie-Tooth (CMT) disease. Recent studies suggest that INF2, through its effects on actin and microtubule arrangement, can regulate processes including vesicle trafficking, cell adhesion, mitochondrial calcium uptake, mitochondrial fission, and T-cell polarization. Despite roles for INF2 in multiple cellular processes, neither the human pathogenic R218Q INF2 point mutation nor the INF2 knock-out allele is sufficient to cause disease in mice. This discrepancy challenges our efforts to explain the disease mechanism, as the link between INF2-related processes, podocyte structure, disease inheritance pattern, and their clinical presentation remains enigmatic. Here, we compared the kidney responses to puromycin aminonucleoside (PAN) induced injury between R218Q INF2 point mutant knock-in and INF2 knock-out mouse models and show that R218Q INF2 mice are susceptible to developing proteinuria and FSGS. This contrasts with INF2 knock-out mice, which show only a minimal kidney phenotype. Co-localization and co-immunoprecipitation analysis of wild-type and mutant INF2 coupled with measurements of cellular actin content revealed that the R218Q INF2 point mutation confers a gain-of-function effect by altering the actin cytoskeleton, facilitated in part by alterations in INF2 localization. Differential analysis of RNA expression in PAN-stressed heterozygous R218Q INF2 point-mutant and heterozygous INF2 knock-out mouse glomeruli showed that the adhesion and mitochondria-related pathways were significantly enriched in the disease condition. Mouse podocytes with R218Q INF2, and an INF2-mutant human patient's kidney organoid-derived podocytes with an S186P INF2 mutation, recapitulate the defective adhesion and mitochondria phenotypes. These results link INF2-regulated cellular processes to the onset and progression of glomerular disease. Thus, our data demonstrate that gain-of-function mechanisms drive INF2-related FSGS and explain the autosomal dominant inheritance pattern of this disease.

2.
Pflugers Arch ; 475(3): 323-341, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36449077

RESUMO

Two heterozygous missense variants (G1 and G2) of Apolipoprotein L1 (APOL1) found in individuals of recent African ancestry can attenuate the severity of infection by some forms of Trypanosoma brucei. However, these two variants within a broader African haplotype also increase the risk of kidney disease in Americans of African descent. Although overexpression of either variant G1 or G2 causes multiple pathogenic changes in cultured cells and transgenic mouse models, the mechanism(s) promoting kidney disease remain unclear. Human serum APOL1 kills trypanosomes through its cation channel activity, and cation channel activity of recombinant APOL1 has been reconstituted in lipid bilayers and proteoliposomes. Although APOL1 overexpression increases whole cell cation currents in HEK-293 cells, the ion channel activity of APOL1 has not been assessed in glomerular podocytes, the major site of APOL1-associated kidney diseases. We characterize APOL1-associated whole cell and on-cell cation currents in HEK-293 T-Rex cells and demonstrate partial inhibition of currents by anti-APOL antibodies. We detect in primary human podocytes a similar cation current inducible by interferon-γ (IFNγ) and sensitive to inhibition by anti-APOL antibody as well as by a fragment of T. brucei Serum Resistance-Associated protein (SRA). CRISPR knockout of APOL1 in human primary podocytes abrogates the IFNγ-induced, antibody-sensitive current. Our novel characterization in HEK-293 cells of heterologous APOL1-associated cation conductance inhibited by anti-APOL antibody and our documentation in primary human glomerular podocytes of endogenous IFNγ-stimulated, APOL1-mediated, SRA and anti-APOL-sensitive ion channel activity together support APOL1-mediated channel activity as a therapeutic target for treatment of APOL1-associated kidney diseases.


Assuntos
Nefropatias , Podócitos , Camundongos , Animais , Humanos , Podócitos/metabolismo , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Células HEK293 , Nefropatias/metabolismo , Camundongos Transgênicos , Canais Iônicos/metabolismo
3.
Front Transplant ; 2: 1342471, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38993907

RESUMO

Introduction: Advances in the field of genetic testing have spurred its use in transplantation. Potential benefits of genetic testing in transplant nephrology include diagnosis, treatment, risk stratification of recurrent disease, and risk stratification in potential donors. However, it is unclear how to best apply genetic testing in this population to maximize its yield. We describe our transplant center's approach to selective genetic testing as part of kidney transplant candidate and donor evaluation. Methods: Transplant recipient candidates were tested if they had a history of ESRD at age <50, primary FSGS, complement-mediated or unknown etiology of kidney disease, or had a family history of kidney disease. Donors were tested if age <35, were related to their potential recipients with known genetic susceptibility or had a first-degree relative with a history of kidney disease of unknown etiology. A targeted NGS gene panel of 385 genes was used. Clinical implications and downstream effects were monitored. Results: Over 30% of recipients tested within the established criteria were positive for a pathogenic variant. The most common pathogenic variants were APOL1 high-risk genotypes as well as collagen 4-alpha-3, -4 and -5. Donor testing done according to our inclusion criteria resulted in about 12% yield. Positive test results in recipients helped with stratification of the risk of recurrent disease. Positive test results in potential donors guided informed decisions on when not to move forward with a donation. Discussion: Integrating targeted panel genetic testing into a kidney transplant clinic in conjunction with a selective criteria for testing donors and recipients ensured a reasonable diagnostic yield. The results had implications on clinical management, risk stratification and in some cases were instrumental in directing downstream changes including when to stop the evaluation process. Given the impact on management and transplant decisions, we advocate for the widespread use of genetic testing in selected individuals undergoing transplant evaluation and donation who meet pre-defined criteria.

4.
Proc Natl Acad Sci U S A ; 119(44): e2210150119, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36282916

RESUMO

APOL1 risk variants are associated with increased risk of kidney disease in patients of African ancestry, but not all individuals with the APOL1 high-risk genotype develop kidney disease. As APOL1 gene expression correlates closely with the degree of kidney cell injury in both cell and animal models, the mechanisms regulating APOL1 expression may be critical determinants of risk allele penetrance. The APOL1 messenger RNA includes Alu elements at the 3' untranslated region that can form a double-stranded RNA structure (Alu-dsRNA) susceptible to posttranscriptional adenosine deaminase acting on RNA (ADAR)-mediated adenosine-to-inosine (A-to-I) editing, potentially impacting gene expression. We studied the effects of ADAR expression and A-to-I editing on APOL1 levels in podocytes, human kidney tissue, and a transgenic APOL1 mouse model. In interferon-γ (IFN-γ)-stimulated human podocytes, ADAR down-regulates APOL1 by preventing melanoma differentiation-associated protein 5 (MDA5) recognition of dsRNA and the subsequent type I interferon (IFN-I) response. Knockdown experiments showed that recognition of APOL1 messenger RNA itself is an important contributor to the MDA5-driven IFN-I response. Mathematical modeling suggests that the IFN-ADAR-APOL1 network functions as an incoherent feed-forward loop, a biological circuit capable of generating fast, transient responses to stimuli. Glomeruli from human kidney biopsies exhibited widespread editing of APOL1 Alu-dsRNA, while the transgenic mouse model closely replicated the edited sites in humans. APOL1 expression in mice was inversely correlated with Adar1 expression under IFN-γ stimuli, supporting the idea that ADAR regulates APOL1 levels in vivo. ADAR-mediated A-to-I editing is an important regulator of APOL1 expression that could impact both penetrance and severity of APOL1-associated kidney disease.


Assuntos
Adenosina Desaminase , Interferon Tipo I , Humanos , Animais , Camundongos , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Edição de RNA , Helicase IFIH1 Induzida por Interferon/metabolismo , RNA de Cadeia Dupla/genética , Regiões 3' não Traduzidas , Apolipoproteína L1/genética , Interferon gama/genética , Interferon gama/metabolismo , RNA Mensageiro/metabolismo , Inosina/genética , Inosina/metabolismo , Adenosina/metabolismo , Interferon Tipo I/metabolismo
5.
J Am Soc Nephrol ; 33(5): 889-907, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35232775

RESUMO

BACKGROUND: Two variants in the gene encoding apolipoprotein L1 (APOL1) that are highly associated with African ancestry are major contributors to the large racial disparity in rates of human kidney disease. We previously demonstrated that recruitment of APOL1 risk variants G1 and G2 from the endoplasmic reticulum to lipid droplets leads to reduced APOL1-mediated cytotoxicity in human podocytes. METHODS: We used CRISPR-Cas9 gene editing of induced pluripotent stem cells to develop human-derived APOL1G0/G0 and APOL1G2/G2 kidney organoids on an isogenic background, and performed bulk RNA sequencing of organoids before and after treatment with IFN-γ. We examined the number and distribution of lipid droplets in response to treatment with inhibitors of diacylglycerol O-acyltransferases 1 and 2 (DGAT1 and DGAT2) in kidney cells and organoids. RESULTS: APOL1 was highly upregulated in response to IFN-γ in human kidney organoids, with greater increases in organoids of high-risk G1 and G2 genotypes compared with wild-type (G0) organoids. RNA sequencing of organoids revealed that high-risk APOL1G2/G2 organoids exhibited downregulation of a number of genes involved in lipogenesis and lipid droplet biogenesis, as well as upregulation of genes involved in fatty acid oxidation. There were fewer lipid droplets in unstimulated high-risk APOL1G2/G2 kidney organoids than in wild-type APOL1G0/G0 organoids. Whereas DGAT1 inhibition reduced kidney organoid lipid droplet number, DGAT2 inhibition unexpectedly increased organoid lipid droplet number. DGAT2 inhibition promoted the recruitment of APOL1 to lipid droplets, with associated reduction in cytotoxicity. CONCLUSIONS: Lipogenesis and lipid droplet formation are important modulators of APOL1-associated cytotoxicity. Inhibition of DGAT2 may offer a potential therapeutic strategy to attenuate cytotoxic effects of APOL1 risk variants.


Assuntos
Nefropatias , Podócitos , Apolipoproteína L1/genética , Diacilglicerol O-Aciltransferase/genética , Feminino , Humanos , Rim , Nefropatias/genética , Gotículas Lipídicas , Masculino
7.
Front Immunol ; 12: 624821, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149688

RESUMO

Kidney disease affects 10% of the world population and is associated with increased mortality. Steroid-resistant nephrotic syndrome (SRNS) is a leading cause of end-stage kidney disease in children, often failing standard immunosuppression. Here, we report the results of a prospective study to investigate the immunological impact and safety of a gluten-free and dairy-free (GF/DF) diet in children with SRNS. The study was organized as a four-week summer camp implementing a strict GF/DF diet with prospective collection of blood, urine and stool in addition to whole exome sequencing WES of DNA of participants. Using flow cytometry, proteomic assays and microbiome metagenomics, we show that GF/DF diet had a major anti-inflammatory effect in all participants both at the protein and cellular level with 4-fold increase in T regulatory/T helper 17 cells ratio and the promotion of a favorable regulatory gut microbiota. Overall, GF/DF can have a significant anti-inflammatory effect in children with SRNS and further trials are warranted to investigate this potential dietary intervention in children with SRNS.


Assuntos
Laticínios/efeitos adversos , Dieta Livre de Glúten , Síndrome Nefrótica/congênito , Adolescente , Biomarcadores/sangue , Biomarcadores/urina , Criança , Pré-Escolar , Citocinas/sangue , Dieta Livre de Glúten/efeitos adversos , Estudos de Viabilidade , Feminino , Microbioma Gastrointestinal , Humanos , Lactente , Mediadores da Inflamação/sangue , Intestinos/microbiologia , Masculino , Síndrome Nefrótica/diagnóstico , Síndrome Nefrótica/dietoterapia , Síndrome Nefrótica/imunologia , Síndrome Nefrótica/microbiologia , Projetos Piloto , Estudo de Prova de Conceito , Estudos Prospectivos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
9.
Kidney Int Rep ; 4(7): 923-929, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31317114

RESUMO

INTRODUCTION: Coding variants in apolipoprotein L-1 (APOL1) are associated with an increased risk of end-stage kidney disease (ESRD) in African American individuals under a recessive model of inheritance. The effect of the APOL1 risk alleles on kidney disease has been observed in studies in African American and African populations. Despite the 130 million individuals of recent African ancestry in South America, the impact of APOL1 has not been explored. METHODS: In this case-control study, we tested APOL1 genotype in 106 Brazilian HD (hemodialysis) patients with African ancestry and compared risk allele frequency with 106 healthy first-degree relatives. The association of risk alleles and ESRD was calculated with a linear mixed model and was adjusted for relatedness and additional confounders. In a broader survey, the age of dialysis initiation and APOL1 variants were analyzed in 274 HD patients. RESULTS: Two APOL1 risk alleles were 10 times more common in patients with ESRD than in controls (9.4% vs. 0.9%; odds ratio [OR]: 10.95, SE = 1.49, P = 0.0017). Carriers of 2 risk alleles initiated dialysis 12 years earlier than patients with zero risk alleles. CONCLUSION: The APOL1 risk variants were less frequent in dialysis patients of African ancestry in Brazil than in the United States. Nonetheless, carriers of 2 risk variants had 10-fold higher odds of ESRD. Age of dialysis initiation was markedly lower in 2-risk allele carriers, suggesting a more aggressive disease phenotype. The Brazilian population represents an opportunity to identify different sets of genetic modifiers or environmental triggers that might be present in more extensively studied populations.

11.
Proc Natl Acad Sci U S A ; 116(9): 3712-3721, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30733285

RESUMO

Two coding variants in the apolipoprotein L1 (APOL1) gene (termed G1 and G2) are strongly associated with increased risk of nondiabetic kidney disease in people of recent African ancestry. The mechanisms by which the risk variants cause kidney damage, although not well-understood, are believed to involve injury to glomerular podocytes. The intracellular localization and function of APOL1 in podocytes remain unclear, with recent studies suggesting possible roles in the endoplasmic reticulum (ER), mitochondria, endosomes, lysosomes, and autophagosomes. Here, we demonstrate that APOL1 also localizes to intracellular lipid droplets (LDs). While a large fraction of risk variant APOL1 (G1 and G2) localizes to the ER, a significant proportion of wild-type APOL1 (G0) localizes to LDs. APOL1 transiently interacts with numerous organelles, including the ER, mitochondria, and endosomes. Treatment of cells that promote LD formation with oleic acid shifted the localization of G1 and G2 from the ER to LDs, with accompanying reduction of autophagic flux and cytotoxicity. Coexpression of G0 APOL1 with risk variant APOL1 enabled recruitment of G1 and G2 from the ER to LDs, accompanied by reduced cell death. The ability of G0 APOL1 to recruit risk variant APOL1 to LDs may help explain the recessive pattern of kidney disease inheritance. These studies establish APOL1 as a bona fide LD-associated protein, and reveal that recruitment of risk variant APOL1 to LDs reduces cell toxicity, autophagic flux, and cell death. Thus, interventions that divert APOL1 risk variants to LDs may serve as a novel therapeutic strategy to alleviate their cytotoxic effects.


Assuntos
Apolipoproteína L1/genética , Autofagia/genética , Nefropatias/genética , Gotículas Lipídicas/metabolismo , População Negra/genética , Retículo Endoplasmático/genética , Endossomos/genética , Variação Genética , Células HEK293 , Humanos , Rim/lesões , Rim/patologia , Nefropatias/fisiopatologia , Gotículas Lipídicas/patologia , Lisossomos/genética , Podócitos/metabolismo , Podócitos/patologia , Fatores de Risco
12.
J. bras. nefrol ; 40(4): 388-402, Out.-Dec. 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-984581

RESUMO

ABSTRACT There are striking differences in chronic kidney disease between Caucasians and African descendants. It was widely accepted that this occurred due to socioeconomic factors, but recent studies show that apolipoprotein L-1 (APOL1) gene variants are strongly associated with focal segmental glomerulosclerosis, HIV-associated nephropathy, hypertensive nephrosclerosis, and lupus nephritis in the African American population. These variants made their way to South America trough intercontinental slave traffic and conferred an evolutionary advantage to the carries by protecting against forms of trypanosomiasis, but at the expense of an increased risk of kidney disease. The effect of the variants does not seem to be related to their serum concentration, but rather to local action on the podocytes. Risk variants are also important in renal transplantation, since grafts from donors with risk variants present worse survival.


RESUMO Existem importantes diferenças na doença renal crônica entre caucasianos e afrodescendentes. Foi amplamente aceito que isso ocorreu devido a fatores socioeconômicos, mas estudos recentes mostraram que as variantes gênicas da apolipoproteína L-1 (APOL1) estão fortemente associadas à glomeruloesclerose segmentar e focal, nefropatia associada ao HIV, nefroesclerose hipertensiva e nefrite lúpica na população afrodescendente. Essas variantes chegaram à América do Sul através do tráfico intercontinental de escravos, e proporcionaram uma vantagem evolutiva aos portadores, protegendo contra formas de tripanossomíase, mas à custa de um maior risco de doença renal. O efeito das variantes não parece estar relacionado à sua concentração sérica, mas sim à sua ação local sobre os podócitos. Variantes de risco também são importantes no transplante renal, já que enxertos de doadores com variantes de risco apresentam pior sobrevida.


Assuntos
Humanos , Insuficiência Renal Crônica/genética , Apolipoproteína L1/genética , Polimorfismo Genético , Variação Genética , Negro ou Afro-Americano/genética , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/epidemiologia , Prevalência , Fatores de Risco , Podócitos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/epidemiologia , Apolipoproteína L1/fisiologia
13.
J Bras Nefrol ; 40(4): 388-402, 2018.
Artigo em Inglês, Português | MEDLINE | ID: mdl-30052698

RESUMO

There are striking differences in chronic kidney disease between Caucasians and African descendants. It was widely accepted that this occurred due to socioeconomic factors, but recent studies show that apolipoprotein L-1 (APOL1) gene variants are strongly associated with focal segmental glomerulosclerosis, HIV-associated nephropathy, hypertensive nephrosclerosis, and lupus nephritis in the African American population. These variants made their way to South America trough intercontinental slave traffic and conferred an evolutionary advantage to the carries by protecting against forms of trypanosomiasis, but at the expense of an increased risk of kidney disease. The effect of the variants does not seem to be related to their serum concentration, but rather to local action on the podocytes. Risk variants are also important in renal transplantation, since grafts from donors with risk variants present worse survival.


Assuntos
Apolipoproteína L1/genética , Insuficiência Renal Crônica/genética , Negro ou Afro-Americano/genética , Apolipoproteína L1/fisiologia , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Variação Genética , Humanos , Podócitos , Polimorfismo Genético , Prevalência , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/etiologia , Fatores de Risco
14.
Nephron Clin Pract ; 128(3-4): 297-302, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25573484

RESUMO

The spectrum of polycystic kidney disease (PKD) comprises a family of inherited syndromes defined by renal cyst formation and growth, progressive renal function loss and variable extrarenal manifestations. The most common form, autosomal-dominant PKD is caused by mutations in one of two genes, PKD1 or PKD2. Recent developments in genomic and proteomic medicine have resulted in the discovery of novel genes implicated in the wide variety of less frequent, recessive PKD syndromes. Cysts are the disease, and overall cystic burden, measured by MRI as total kidney volume, is being established as the best available biomarker of disease progression. Current state-of-the-art therapy is aimed at quality treatment for chronic renal insufficiency and cyst-related complications. Recent therapeutic studies have focused on mechanisms reducing intracellular cyclic AMP levels, blocking the renin-angiotensin-aldosterone system and inhibiting the mTOR-signaling pathway. PKD therapies with vasopressin antagonists and somatostatin analogues result in the reduction of intracellular cAMP levels and have shown limited clinical success, but side effects are prominent. Similarly, mTOR pathway inhibition has not shown significant therapeutic benefits. While the HALT-PKD study will answer questions by the end of 2014 about the utility of renin-angiotensin-aldosterone system blockade and aggressive blood pressure control, the next generation of PKD therapy studies targeting proliferative mechanisms of cyst expansion are already under way. Advances in research on the molecular mechanisms of cystogenesis will help design novel targeted PKD therapies in the future.


Assuntos
Doenças Renais Policísticas/tratamento farmacológico , Biomarcadores/análise , AMP Cíclico/antagonistas & inibidores , Progressão da Doença , Genômica , Humanos , Mutação , Doenças Renais Policísticas/genética , Proteômica , Sistema Renina-Angiotensina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Canais de Cátion TRPP/genética
15.
Nephrol Dial Transplant ; 28(2): 438-46, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23291370

RESUMO

BACKGROUND: Markers of inflammation are linked to malnutrition and confer an increased mortality risk in hemodialysis patients. Ultrapure dialysate might have a beneficial effect on markers of inflammation. We conducted a meta-analysis that examined the effect of ultrapure versus standard dialysate on markers of inflammation, oxidative stress, nutrition and anemia parameters. METHODS: We performed a literature search using MEDLINE, Scopus, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov and scientific abstracts. Single-arm studies, nonrandomized and randomized controlled trials were included. We conducted random effects model meta-analyses to assess changes in the aforementioned outcomes. RESULTS: We identified 16 single-arm studies, 2 crossover and 3 parallel-arm nonrandomized controlled trials and 5 crossover and 5 parallel-arm randomized controlled trials. In an analysis of 23 study arms or cohorts (n = 2221), ultrapure dialysate resulted in a significant decrease in C-reactive protein (-3.2 mg/L; 95% CI -4.6, -1.8; P < 0.001). Other markers of inflammation and oxidative stress displayed similar significant improvements. Ultrapure dialysate also resulted in a significant increase in serum albumin (0.11 g/dL; 95% CI 0.02, 0.19; P = 0.011) and hemoglobin (0.40 g/dL; 95% CI 0.06, 0.75; P = 0.022), and a decrease in the weekly erythropoietin dose (-273 units; 95% CI -420, -126; P < 0.001). The results remained significant in analyses restricted to controlled trials. CONCLUSIONS: Use of ultrapure dialysate in hemodialysis patients results in a decrease in markers of inflammation and oxidative stress, an increase in serum albumin and hemoglobin and a decrease in erythropoietin requirement. Although improvement in these surrogate endpoints might confer a cardiovascular benefit, a large trial with hard clinical endpoints is required.


Assuntos
Anemia/metabolismo , Soluções para Diálise/farmacologia , Inflamação/metabolismo , Estado Nutricional/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Diálise Renal/métodos , Idoso , Biomarcadores/metabolismo , Proteína C-Reativa/metabolismo , Soluções para Diálise/normas , Feminino , Hemoglobinas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Estado Nutricional/fisiologia , Estresse Oxidativo/fisiologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA