Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33536341

RESUMO

Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl-/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.


Assuntos
Anidrase Carbônica II/genética , Fatores de Transcrição Forkhead/genética , Insuficiência Renal/genética , Proteína 1 do Complexo Esclerose Tuberosa/genética , Animais , Cistos/genética , Cistos/patologia , Modelos Animais de Doenças , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Mutação/genética , ATPases Translocadoras de Prótons/genética , Insuficiência Renal/patologia , Canais de Cátion TRPP/genética , Esclerose Tuberosa
2.
Int J Mol Sci ; 21(5)2020 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-32138326

RESUMO

The tuberous sclerosis complex (Tsc) proteins regulate the conserved mTORC1 growth regulation pathway. We identified that loss of the Tsc2 gene in mouse inner medullary collecting duct (mIMCD) cells induced a greater than two-fold increase in extracellular vesicle (EV) production compared to the same cells having an intact Tsc axis. We optimized EV isolation using a well-established size exclusion chromatography method to produce high purity EVs. Electron microscopy confirmed the purity and spherical shape of EVs. Both tunable resistive pulse sensing (TRPS) and dynamic light scattering (DLS) demonstrated that the isolated EVs possessed a heterogenous size distribution. Approximately 90% of the EVs were in the 100-250 nm size range, while approximately 10% had a size greater than 250 nm. Western blot analysis using proteins isolated from the EVs revealed the cellular proteins Alix and TSG101, the transmembrane proteins CD63, CD81, and CD9, and the primary cilia Hedgehog signaling-related protein Arl13b. Proteomic analysis of EVs identified a significant difference between the Tsc2-intact and Tsc2-deleted cell that correlated well with the increased production. The EVs may be involved in tissue homeostasis and cause disease by overproduction and altered protein content. The EVs released by renal cyst epithelia in TSC complex may serve as a tool to discover the mechanism of TSC cystogenesis and in developing potential therapeutic strategies.


Assuntos
Vesículas Extracelulares/metabolismo , Rim/metabolismo , Esclerose Tuberosa/metabolismo , Animais , Western Blotting , Linhagem Celular , Cromatografia em Gel , Vesículas Extracelulares/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Ligação Proteica , Proteômica , Tetraspanina 28/metabolismo , Tetraspanina 29/metabolismo , Tetraspanina 30/metabolismo , Esclerose Tuberosa/genética
3.
Nephrol Dial Transplant ; 34(6): 1000-1008, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30053159

RESUMO

BACKGROUND: A reduction in renal angiomyolipoma volume observed with everolimus (EVE) treatment in patients with tuberous sclerosis complex (TSC) has been postulated to translate to clinical benefit by reducing the risk of renal hemorrhage and chronic renal failure. METHODS: The long-term effects of EVE on renal function (∼4 years of treatment) were examined in patients treated with EVE in the Phase 3 EXIST-1 and EXIST-2 studies. Patients in EXIST-1 had TSC and subependymal giant cell astrocytoma (SEGA), and patients in EXIST-2 had renal angiomyolipoma and a definite diagnosis of TSC or sporadic lymphangioleiomyomatosis. EVE was administered at 4.5 mg/m2/day, with adjustment to achieve target trough levels of 5-15 ng/mL in EXIST-1 and at 10 mg/day in EXIST-2. Estimated glomerular filtration rate (eGFR) and creatinine levels were assessed at baseline, at Weeks 2, 4, 6, 8, 12 and 18, then every 3 months thereafter. Proteinuria was graded according to National Cancer Institute Common Terminology Criteria for Adverse Events version 3.0. RESULTS: A total of 111 patients from EXIST-1 and 112 patients from EXIST-2 were included in this analysis. Respective mean ages at EVE initiation were 10.5 [standard deviation (SD) 6.45] and 33.2 (SD 10.29) years, and 3.6% and 37.5% of patients had undergone prior renal intervention. Mean baseline eGFR was 115 and 88 mL/min/1.73 m2 in EXIST-1 and EXIST-2, respectively. Overall, mean eGFR remained stable over time in both studies, with an decline in renal function mostly confined to some patients with severely compromised renal function before treatment. Patients with prior renal intervention exhibited low eGFR values throughout the study. The incidence of proteinuria increased after initiating treatment with EVE and was mostly Grade 1/2 in severity, with Grade 3 proteinuria reported in only two patients. Measurements of proteinuria were limited by the use of urine dipstick tests. CONCLUSIONS: The use of EVE does not appear to be nephrotoxic in patients with SEGA or renal angiomyolipoma associated with TSC and may preserve renal function in most patients.ClinicalTrials.gov identifiers NCT00789828 and NCT00790400.


Assuntos
Angiomiolipoma/tratamento farmacológico , Everolimo/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Esclerose Tuberosa/tratamento farmacológico , Adolescente , Adulto , Antineoplásicos/uso terapêutico , Astrocitoma , Criança , Pré-Escolar , Método Duplo-Cego , Feminino , Taxa de Filtração Glomerular , Humanos , Imunossupressores/uso terapêutico , Lactente , Rim , Falência Renal Crônica/complicações , Linfangioleiomiomatose/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Proteinúria/tratamento farmacológico , Insuficiência Renal Crônica/complicações , Estudos Retrospectivos , Adulto Jovem
4.
Am J Med Genet C Semin Med Genet ; 178(3): 338-347, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30307110

RESUMO

Tuberous sclerosis complex (TSC) is a tumor predisposition syndrome with significant renal cystic and solid tumor disease. It commonly causes several types of cystic disease and benign tumors (angiomyolipomata) in the kidneys that can both lead to significant premature loss of glomerular filtration rate. The main risks of angiomyolipomata, severe bleeding, loss of renal function, and pulmonary lymphangioleiomyomatosis, can be ameliorated by active surveillance and preemptive therapy with mTOR inhibitors. The cystogenic mechanism may involve primary cilia, but also appears to also involve a majority of normal tubular cells and may be driven by a minority of cells with mutations inactivating both their TSC1 or TSC2 genes. Malignant tumors are rare.


Assuntos
Neoplasias Renais/etiologia , Insuficiência Renal Crônica/terapia , Esclerose Tuberosa/etiologia , Angiomiolipoma/etiologia , Animais , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Doenças Renais Císticas/etiologia , Linfangioleiomiomatose/etiologia , Mutação , Insuficiência Renal Crônica/etiologia , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/genética
5.
Pediatr Nephrol ; 33(1): 101-109, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28993887

RESUMO

BACKGROUND: Patients with tuberous sclerosis complex (TSC) often have multiple TSC-associated hamartomas, particularly in the brain and kidney. METHODS: This was a post hoc analysis of pediatric patients being treated for subependymal giant cell astrocytomas (SEGAs) during the phase 3, randomized, double-blind, placebo-controlled EXIST-1 trial. Patients were initially randomly assigned to receive everolimus 4.5 mg/m2/day (target blood trough 5-15 mg/dl) or placebo and could continue in an open-label extension phase. Angiomyolipoma response rates were analyzed in patients aged <18 years with ≥1 target angiomyolipoma lesion at baseline. Response was defined as the proportion of patients with a ≥50% reduction in the sum volume of target renal angiomyolipomata from baseline, in the absence of new target angiomyolipomata, a >20% increase in kidney volume from nadir, and angiomyolipoma-related bleeding ≥ grade 2. Tolerability was also assessed. RESULTS: Overall, this analysis included 33 patients. Renal angiomyolipoma response was achieved by 75.8% of patients (95% confidence interval, 57.7-88.9%), with sustained mean reductions in renal angiomyolipoma volume over nearly 4 years of treatment. In addition, most (≥80%) achieved clinically relevant reductions in angiomyolipoma volume (≥50%), beginning at week 24 and continuing for the remainder of the study. Everolimus was generally well tolerated in this subgroup, with most adverse events being grade 1 or 2 in severity. CONCLUSIONS: Although everolimus is currently not indicated for this use, this analysis from EXIST-1 demonstrates its long-term efficacy and safety for the treatment of renal angiomyolipoma in pediatric patients undergoing treatment for TSC-associated SEGA.


Assuntos
Angiomiolipoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Everolimo/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Esclerose Tuberosa/complicações , Adolescente , Angiomiolipoma/complicações , Antineoplásicos/efeitos adversos , Astrocitoma/complicações , Astrocitoma/tratamento farmacológico , Criança , Pré-Escolar , Método Duplo-Cego , Everolimo/efeitos adversos , Feminino , Humanos , Rim/patologia , Neoplasias Renais/patologia , Masculino , Resultado do Tratamento , Esclerose Tuberosa/tratamento farmacológico
6.
Am J Physiol Renal Physiol ; 312(4): F791-F805, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28122715

RESUMO

Primary cilia sense environmental conditions, including osmolality, but whether cilia participate in the osmotic response in renal epithelial cells is not known. The transient receptor potential (TRP) channels TRPV4 and TRPM3 are osmoresponsive. TRPV4 localizes to cilia in certain cell types, while renal subcellular localization of TRPM3 is not known. We hypothesized that primary cilia are required for maximal activation of the osmotic response of renal epithelial cells and that ciliary TRPM3 and TRPV4 mediate that response. Ciliated [murine epithelial cells from the renal inner medullary collecting duct (mIMCD-3) and 176-5] and nonciliated (176-5Δ) renal cells expressed Trpv4 and Trpm3 Ciliary expression of TRPM3 was observed in mIMCD-3 and 176-5 cells and in wild-type mouse kidney tissue. TRPV4 was identified in cilia and apical membrane of mIMCD-3 cells by electrophysiology and in the cell body by immunofluorescence. Hyperosmolal stress at 500 mOsm/kg (via NaCl addition) induced the osmotic response genes betaine/GABA transporter (Bgt1) and aldose reductase (Akr1b3) in all ciliated cell lines. This induction was attenuated in nonciliated cells. A TRPV4 agonist abrogated Bgt1 and Akr1b3 induction in ciliated and nonciliated cells. A TRPM3 agonist attenuated Bgt1 and Akr1b3 induction in ciliated cells only. TRPM3 knockout attenuated Akr1b3 induction. Viability under osmotic stress was greater in ciliated than nonciliated cells. Akr1b3 induction was also less in nonciliated than ciliated cells when mannitol was used to induce hyperosmolal stress. These findings suggest that primary cilia are required for the maximal osmotic response in renal epithelial cells and that TRPM3 is involved in this mechanism. TRPV4 appears to modulate the osmotic response independent of cilia.


Assuntos
Células Epiteliais/metabolismo , Túbulos Renais Coletores/metabolismo , Osmorregulação , Pressão Osmótica , Canais de Cátion TRPM/metabolismo , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Cílios/metabolismo , Células Epiteliais/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Edição de Genes , Hidroxiprostaglandina Desidrogenases/genética , Hidroxiprostaglandina Desidrogenases/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osmorregulação/efeitos dos fármacos , Pressão Osmótica/efeitos dos fármacos , Solução Salina Hipertônica/farmacologia , Transdução de Sinais , Canais de Cátion TRPM/genética , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Transfecção
7.
Am J Respir Crit Care Med ; 194(6): 748-61, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27628078

RESUMO

BACKGROUND: Lymphangioleiomyomatosis (LAM) is a rare cystic lung disease that primarily affects women. The purpose of these guidelines is to provide recommendations for the diagnosis and treatment of LAM. METHODS: Systematic reviews were performed to summarize evidence pertinent to our questions. The evidence was summarized and discussed by a multidisciplinary panel. Evidence-based recommendations were then formulated, written, and graded using the Grading of Recommendations, Assessment, Development, and Evaluation approach. RESULTS: After considering the panel's confidence in the estimated effects, the balance of desirable (i.e., benefits) and undesirable (i.e., harms and burdens) consequences of treatment, patient values and preferences, cost, and feasibility, recommendations were formulated for or against specific interventions. These included recommendations for sirolimus treatment and vascular endothelial growth factor D testing and recommendations against doxycycline and hormonal therapy. CONCLUSIONS: Evidence-based recommendations for the diagnosis and treatment of patients with LAM are provided. Frequent reassessment and updating will be needed.


Assuntos
Linfangioleiomiomatose/diagnóstico , Biópsia , Feminino , Humanos , Pulmão/diagnóstico por imagem , Pulmão/patologia , Pulmão/fisiopatologia , Linfangioleiomiomatose/fisiopatologia , Linfangioleiomiomatose/terapia , Masculino , Sirolimo/uso terapêutico , Tomografia Computadorizada por Raios X , Fator D de Crescimento do Endotélio Vascular/sangue
8.
Nephrol Dial Transplant ; 31(1): 111-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26156073

RESUMO

BACKGROUND: Mammalian target of rapamycin (mTOR) inhibitors are recommended as first-line treatment of renal angiomyolipoma associated with tuberous sclerosis complex (TSC) or sporadic lymphangioleiomyomatosis (sporadic LAM), but follow-up is limited. Longer term efficacy and tolerability data from a Phase 3, double-blind, placebo-controlled trial are presented. METHODS: Following favorable results from the primary analysis (data cutoff 30 June 2011) of the EXIST-2 trial, patients still receiving study treatment were allowed to enter an open-label extension. Everolimus was initiated at 10 mg once daily and titrated based on tolerability. The primary outcome was angiomyolipoma response rate (≥ 50% reduction from baseline in target lesion volumes). Safety was a secondary endpoint. RESULTS: As of the cutoff date (1 May 2013), 112 patients had received everolimus, and the response rate in 107 patients with angiomyolipoma (median duration of medication exposure of 28.9 months) was 54%. The proportion of patients achieving angiomyolipoma reductions of ≥ 30% and ≥ 50% increased over time, reaching 81.6% (62/76) and 64.5% (49/76), respectively, by Week 96. No everolimus-treated patients experienced renal bleeding. The long-term safety profile was consistent with previous reports; adverse events (AEs) were mostly Grade 1/2, and there were no new safety issues. The frequency of emerging AEs and severe AEs lessened over time. CONCLUSIONS: Longer term everolimus treatment appeared safe and effective in patients with TSC- or sporadic LAM-associated renal angiomyolipoma not requiring surgical intervention. Continued reduction in angiomyolipoma volume was demonstrated, and there was no angiomyolipoma-related bleeding; AEs were predictable and generally manageable. TRIAL REGISTRATION: clinicaltrialsgov identifier: NCT00790400 (http://clinicaltrials.gov/ct2/show/NCT00790400).


Assuntos
Angiomiolipoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Everolimo/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Linfangioleiomiomatose/tratamento farmacológico , Esclerose Tuberosa/tratamento farmacológico , Adolescente , Adulto , Angiomiolipoma/patologia , Intervalo Livre de Doença , Método Duplo-Cego , Feminino , Humanos , Estimativa de Kaplan-Meier , Rim/patologia , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Adulto Jovem
9.
Br J Clin Pharmacol ; 81(5): 958-70, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26580489

RESUMO

AIMS: The purpose was to determine the exposure-response relationship of everolimus in patients with angiomyolipoma from the EXIST-2 trial and to analyze the correlation between exposure and plasma concentrations of angiogenic biomarkers in these patients. METHODS: One hundred and eighteen patients with angiomyolipoma associated with tuberous sclerosis complex (TSC) or sporadic lymphangioleiomyomatosis (sLAM) were randomly assigned 2 : 1 to receive everolimus 10 mg (n = 79) or placebo (n = 39) once daily. Blood samples for determining everolimus concentration were collected at weeks 2, 4, 12, 24 and 48 during double-blind treatment. Plasma samples for biomarker analysis were collected at baseline and weeks 4, 12, 24, 36, 48 and at the end of treatment. Concentrations of eight angiogenic biomarkers associated with tumour growth were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: Peak and trough concentrations of everolimus in blood remained stable over time and similar to those reported in other indications. Substantial pharmacodynamic effects were observed in the everolimus, but not placebo, arm for three biomarkers: After 24 weeks of treatment, reduction of vascular endothelial growth factor D (VEGF-D) and collagen type IV (COL-IV) (mean fold-changes with 95% confidence intervals [CI] were 0.36 [0.33, 0.40], and 0.54 [0.51, 0.57], respectively, P < 0.001 for both), along with increased VEGF-A (mean fold-change of 1.59 [1.39, 1.80], P < 0.001), were seen. Furthermore, baseline VEGF-D and COL-IV levels were associated with angiomyolipoma size at baseline and with angiomyolipoma response to everolimus. CONCLUSIONS: These findings suggest that plasma angiogenic markers may provide an objective measure of patient response to everolimus.


Assuntos
Angiomiolipoma/tratamento farmacológico , Antineoplásicos/farmacologia , Everolimo/farmacologia , Neoplasias Renais/tratamento farmacológico , Linfangioleiomiomatose/tratamento farmacológico , Esclerose Tuberosa/tratamento farmacológico , Adulto , Angiomiolipoma/complicações , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Colágeno Tipo IV/sangue , Relação Dose-Resposta a Droga , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , Everolimo/farmacocinética , Everolimo/uso terapêutico , Humanos , Neoplasias Renais/complicações , Linfangioleiomiomatose/complicações , Esclerose Tuberosa/complicações , Fator A de Crescimento do Endotélio Vascular/sangue , Fator D de Crescimento do Endotélio Vascular/sangue
10.
Am J Physiol Renal Physiol ; 309(1): F79-87, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25904703

RESUMO

In polycystic kidney disease (PKD), the rate of cyst formation and disease progression is highly variable. The lack of predictability in disease progression may be due to additional environmental factors or pathophysiological processes called "third hits." Diabetes is a growing epidemic, and recent studies suggest that PKD patients may be at an increased risk for this disease. We sought to determine if hyperglycemia enhances the initiation and rate of cystogenesis. Tamoxifen was administered to adult Ift88 conditional floxed allele mice to induce cilia loss in the presence of Cre. Subsequent administration of streptozotocin resulted in equivalent hyperglycemia in cilia(+) and cilia(-) mice. Hyperglycemia with loss of cilia increased the rate of cyst formation and cell proliferation. Structural and functional alterations in the kidney, including focal glomerular foot process effacement, interstitial inflammation, formation of primitive renal tubules, polyuria, and increased proteinuria, were also observed in hyperglycemic cilia(-) mice. Gene array analysis indicated enhanced Wnt and epithelial-to-mesenchymal transition signaling in the kidney of hyperglycemic cilia(-) mice. These data show that hyperglycemia, in the absence of cilia, results in renal structural and functional damage and accelerates cystogenesis, suggesting that diabetes is a risk factor in the progression of PKD.


Assuntos
Hiperglicemia/complicações , Rim/patologia , Doenças Renais Policísticas/etiologia , Animais , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Hemodinâmica , Hiperglicemia/patologia , Hiperglicemia/fisiopatologia , Testes de Função Renal , Masculino , Camundongos Knockout , Doenças Renais Policísticas/patologia , Distribuição Aleatória , Proteínas Wnt/metabolismo
11.
BMC Med Genet ; 16: 39, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-26077033

RESUMO

BACKGROUND: Mutations in TSC1 or TSC2 cause the tuberous sclerosis complex (TSC), while mutations in PKD1 or PKD2 cause autosomal dominant polycystic kidney disease (ADPKD). PKD1 lays immediately adjacent to TSC2 and deletions involving both genes, the PKD1/TSC2 contiguous gene syndrome (CGS), are characterized by severe ADPKD, plus TSC. mTOR inhibitors have proven effective in reducing angiomyolipoma (AML) in TSC and total kidney volume in ADPKD but without a positive effect on renal function. METHODS AND RESULTS: We describe a patient with independent truncating PKD1 and TSC2 mutations who has the expected phenotype for both diseases independently instead of the severe one described in PKD1/TSC2-CGS. Treatment with mTOR inhibitors reduced the AML and kidney volume for 2 years but thereafter they resumed growth; no positive effect on renal function was seen throughout. This is the first case addressing the response to mTOR treatment when independent truncating mutations in PKD1 and TSC2 are present. CONCLUSIONS: This case reveals that although PKD1 and TSC2 are adjacent genes and there is likely cross-talk between the PKD1 and TSC2 signalling pathways regulating mTOR, having independent TSC2 and PKD1 mutations can give rise to a milder kidney phenotype than is typical in PKD1/TSC2-CGS cases. A short-term beneficial effect of mTOR inhibition on AML and total kidney volume was not reflected in improved renal function.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Canais de Cátion TRPP/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Fenótipo , Inibidores de Proteínas Quinases/uso terapêutico , Resultado do Tratamento , Esclerose Tuberosa/diagnóstico , Esclerose Tuberosa/tratamento farmacológico , Esclerose Tuberosa/genética , Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa
12.
Curr Opin Pediatr ; 27(2): 227-32, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25689458

RESUMO

PURPOSE OF REVIEW: All polycystic diseases of the kidney exhibit tubular or saccular cysts. The cysts can either be open to the tubule or isolated sacs that have lost their connections. Polycystic kidney diseases derived from different genetic mutations share basic mechanisms of cytogenesis, formation, and progressive enlargement, involving a cellular organelle called the primary cilium. Given the mechanistic commonalities, this review will focus on the therapeutic approaches currently available or under development that likely apply to all inherited renal cystic diseases. RECENT FINDINGS: Recent advances in clinical trials and preclinical experiments have illuminated common signaling pathway involvement. SUMMARY: Avoidance of nephrotoxic drugs or radio-contrast and maintaining normal BMI are routine preventive measures. Limiting the intake of calories, salt, and protein, together with increased intake of fruits, vegetables, and water are dietary treatments that should be started early in the course of the disease. Potential pharmacological treatments targeting cyst initiation and progression are on the horizon.


Assuntos
Dieta Hipossódica , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Doenças Renais Policísticas/terapia , Pravastatina/uso terapêutico , Sistema Renina-Angiotensina/efeitos dos fármacos , Aconselhamento Diretivo , Progressão da Doença , Exercício Físico , Frutas , Humanos , Doenças Renais Policísticas/fisiopatologia , Prognóstico , Comportamento de Redução do Risco , Verduras
13.
J Am Soc Nephrol ; 25(6): 1187-97, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24511141

RESUMO

Domoic acid (DA), an excitatory amino acid produced by diatoms belonging to the genus Pseudo-nitzschia, is a glutamate analog responsible for the neurologic condition referred to as amnesic shellfish poisoning. To date, the renal effects of DA have been underappreciated, although renal filtration is the primary route of systemic elimination and the kidney expresses ionotropic glutamate receptors. To characterize the renal effects of DA, we administered either a neurotoxic dose of DA or doses below the recognized limit of toxicity to adult Sv128/Black Swiss mice. DA preferentially accumulated in the kidney and elicited marked renal vascular and tubular damage consistent with acute tubular necrosis, apoptosis, and renal tubular cell desquamation, with toxic vacuolization and mitochondrial swelling as hallmarks of the cellular damage. Doses≥0.1 mg/kg DA elevated the renal injury biomarkers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, and doses≥0.005 mg/kg induced the early response genes c-fos and junb. Coadministration of DA with the broad spectrum excitatory amino acid antagonist kynurenic acid inhibited induction of c-fos, junb, and neutrophil gelatinase-associated lipocalin. These findings suggest that the kidney may be susceptible to excitotoxic agonists, and renal effects should be considered when examining glutamate receptor activation. Additionally, these results indicate that DA is a potent nephrotoxicant, and potential renal toxicity may require consideration when determining safe levels for human exposure.


Assuntos
Ácido Caínico/análogos & derivados , Toxinas Marinhas/toxicidade , Fármacos Neuromusculares Despolarizantes/toxicidade , Junção Neuromuscular/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Ácido Caínico/farmacocinética , Ácido Caínico/toxicidade , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Toxinas Marinhas/farmacocinética , Camundongos Endogâmicos , Microscopia Eletrônica de Transmissão , Dilatação Mitocondrial/efeitos dos fármacos , Miocárdio/metabolismo , Miocárdio/patologia , Fármacos Neuromusculares Despolarizantes/farmacocinética , Junção Neuromuscular/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vacúolos/patologia , Vacúolos/ultraestrutura , Receptor de GluK2 Cainato
14.
Am J Physiol Renal Physiol ; 307(5): F560-70, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24920756

RESUMO

Nearly all patients with tuberous sclerosis complex (TSC) develop renal angiomyolipomas, although the tumor cell of origin is unknown. We observed decreased renal angiomyolipoma development in patients with TSC2- polycystic kidney disease 1 deletion syndrome and hypertension that were treated from an early age with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers compared with patients who did not receive this therapy. TSC-associated renal angiomyolipomas expressed ANG II type 1 receptors, platelet-derived growth factor receptor-ß, desmin, α-smooth muscle actin, and VEGF receptor 2 but did not express the adipocyte marker S100 or the endothelial marker CD31. Sera of TSC patients exhibited increased vascular mural cell-secreted peptides, such as VEGF-A, VEGF-D, soluble VEGF receptor 2, and collagen type IV. These findings suggest that angiomyolipomas may arise from renal pericytes. ANG II treatment of angiomyolipoma cells in vitro resulted in an exaggerated intracellular Ca(2+) response and increased proliferation, which were blocked by the ANG II type 2 receptor antagonist valsartan. Blockade of ANG II signaling may have preventative therapeutic potential for angiomyolipomas.


Assuntos
Angiomiolipoma/tratamento farmacológico , Angiomiolipoma/patologia , Antagonistas de Receptores de Angiotensina/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Pericitos/patologia , Esclerose Tuberosa/complicações , Angiomiolipoma/fisiopatologia , Angiotensina II/fisiologia , Antagonistas de Receptores de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Humanos , Técnicas In Vitro , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Neoplasias Renais/fisiopatologia , Receptor Tipo 1 de Angiotensina/fisiologia , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Tetrazóis/farmacologia , Tetrazóis/uso terapêutico , Esclerose Tuberosa/patologia , Esclerose Tuberosa/fisiopatologia , Valina/análogos & derivados , Valina/farmacologia , Valina/uso terapêutico , Valsartana
15.
Lancet ; 381(9869): 817-24, 2013 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-23312829

RESUMO

BACKGROUND: Angiomyolipomas are slow-growing tumours associated with constitutive activation of mammalian target of rapamycin (mTOR), and are common in patients with tuberous sclerosis complex and sporadic lymphangioleiomyomatosis. The insidious growth of these tumours predisposes patients to serious complications including retroperitoneal haemorrhage and impaired renal function. Everolimus, a rapamycin derivative, inhibits the mTOR pathway by acting on the mTOR complex 1. We compared the angiomyolipoma response rate on everolimus with placebo in patients with tuberous sclerosis or sporadic lymphanioleiomyomatosis-associated angiomyolipomata. METHODS: In this double-blind, placebo-controlled, phase 3 trial, patients aged 18 years or older with at least one angiomyolipoma 3 cm or larger in its longest diameter (defined by radiological assessment) and a definite diagnosis of tuberous sclerosis or sporadic lymphangioleiomyomatosis were randomly assigned, in a 2:1 fashion with the use of an interactive web response system, to receive oral everolimus 10 mg per day or placebo. The primary efficacy endpoint was the proportion of patients with confirmed angiomyolipoma response of at least a 50% reduction in total volume of target angiomyolipomas relative to baseline. This study is registered with ClinicalTrials.gov number NCT00790400. RESULTS: 118 patients (median age 31·0 years; IQR 18·0­61·0) from 24 centres in 11 countries were randomly assigned to receive everolimus (n=79) or placebo (n=39). At the data cutoff, double-blind treatment was ongoing for 98 patients; two main reasons for discontination were disease progression (nine placebo patients) followed by adverse events (two everolimus patients; four placebo patients). The angiomyolipoma response rate was 42% (33 of 79 [95% CI 31­53%]) for everolimus and 0% (0 of 39 [0­9%]) for placebo (response rate difference 42% [24­58%]; one-sided Cochran-Mantel-Haenszel test p<0·0001). The most common adverse events in the everolimus and placebo groups were stomatitis (48% [38 of 79], 8% [3 of 39], respectively), nasopharyngitis (24% [19 of 79] and 31% [12 of 39]), and acne-like skin lesions (22% [17 of 79] and 5% [2 of 39]). INTERPRETATION: Everolimus reduced angiomyolipoma volume with an acceptable safety profile, suggesting it could be a potential treatment for angiomyolipomas associated with tuberous sclerosis. FUNDING: Novartis Pharmaceuticals.


Assuntos
Angiomiolipoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Sirolimo/análogos & derivados , Adulto , Angiomiolipoma/complicações , Método Duplo-Cego , Everolimo , Feminino , Humanos , Linfangioleiomiomatose/complicações , Masculino , Estudos Prospectivos , Sirolimo/uso terapêutico , Esclerose Tuberosa/complicações
16.
Nephrol Dial Transplant ; 29(6): 1203-10, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24729041

RESUMO

BACKGROUND: Tuberous sclerosis complex (TSC) is characterized by benign tumours in multiple organs, including the brain, kidneys, skin, lungs and heart. Our objective was to evaluate everolimus, an mTOR inhibitor, in the treatment of angiomyolipoma in patients with subependymal giant cell astrocytoma (SEGA) associated with TSC. METHODS: EXamining everolimus In a Study of Tuberous Sclerosis Complex-1 (NCT00789828), a prospective, double-blind, randomized, placebo-controlled, Phase 3 study, examined everolimus in treating SEGA associated with TSC. Patients with serial SEGA growth from pre-baseline to baseline scans were randomly assigned (2:1) to receive 4.5 mg/m(2)/day everolimus (target blood trough: 5-15 ng/mL; n = 78) or placebo (n = 39). Angiomyolipoma response rates were analysed in patients (n = 44) with target baseline angiomyolipoma lesions (≥1 angiomyolipoma; longest diameter ≥1.0 cm). An angiomyolipoma response rate, defined as the proportion of patients with confirmed angiomyolipoma response, was assessed by kidney CT or MRI screening at baseline, at 12, 24 and 48 weeks and annually. RESULTS: Angiomyolipoma response rates were 53.3% (16/30) and 0% (0/14) for everolimus- and placebo-treated patients, respectively. Angiomyolipoma reductions ≥50% in the sum of volumes of all target lesions were seen only in everolimus-treated patients (56.5, 78.3 and 80.0%) compared with placebo-treated patients (0% at each time point) at Weeks 12, 24 and 48, respectively. Greater percentages of everolimus-treated patients had angiomyolipoma reductions ≥30% at these same time points (82.6, 100 and 100% versus 8.3, 18.2 and 16.7% for everolimus versus placebo, respectively). CONCLUSIONS: Everolimus showed efficacy in reducing angiomyolipoma lesion volume in patients with SEGA associated with TSC.The trial is registered with ClinicalTrials.gov, number NCT00789828; http://clinicaltrials.gov/ct2/show/NCT00789828?term=EXIST-1&rank=1.


Assuntos
Angiomiolipoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Astrocitoma/etiologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Primárias Múltiplas/tratamento farmacológico , Sirolimo/análogos & derivados , Esclerose Tuberosa/complicações , Adulto , Astrocitoma/epidemiologia , Encéfalo , Pré-Escolar , Método Duplo-Cego , Everolimo , Feminino , Humanos , Neoplasias Renais/complicações , Masculino , Estudos Prospectivos , Inibidores de Proteínas Quinases/uso terapêutico , Sirolimo/uso terapêutico , Resultado do Tratamento , Esclerose Tuberosa/epidemiologia
17.
J Bras Nefrol ; 46(3): e20240013, 2024.
Artigo em Inglês, Português | MEDLINE | ID: mdl-38991206

RESUMO

Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by the development of hamartomas in the central nervous system, heart, skin, lungs, and kidneys and other manifestations including seizures, cortical tubers, radial migration lines, autism and cognitive disability. The disease is associated with pathogenic variants in the TSC1 or TSC2 genes, resulting in the hyperactivation of the mTOR pathway, a key regulator of cell growth and metabolism. Consequently, the hyperactivation of the mTOR pathway leads to abnormal tissue proliferation and the development of solid tumors. Kidney involvement in TSC is characterized by the development of cystic lesions, renal cell carcinoma and renal angiomyolipomas, which may progress and cause pain, bleeding, and loss of kidney function. Over the past years, there has been a notable shift in the therapeutic approach to TSC, particularly in addressing renal manifestations. mTOR inhibitors have emerged as the primary therapeutic option, whereas surgical interventions like nephrectomy and embolization being reserved primarily for complications unresponsive to clinical treatment, such as severe renal hemorrhage. This review focuses on the main clinical characteristics of TSC, the mechanisms underlying kidney involvement, the recent advances in therapy for kidney lesions, and the future perspectives.


Assuntos
Esclerose Tuberosa , Esclerose Tuberosa/complicações , Esclerose Tuberosa/genética , Esclerose Tuberosa/terapia , Humanos , Neoplasias Renais/terapia , Neoplasias Renais/etiologia , Inibidores de MTOR/uso terapêutico , Serina-Treonina Quinases TOR , Angiomiolipoma/etiologia , Angiomiolipoma/terapia , Nefrologia , Proteína 1 do Complexo Esclerose Tuberosa/genética , Carcinoma de Células Renais/etiologia , Carcinoma de Células Renais/terapia , Carcinoma de Células Renais/genética
18.
J Biol Chem ; 287(40): 33412-23, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22872635

RESUMO

DNA sequences prone to forming noncanonical structures (hairpins, triplexes, G-quadruplexes) cause DNA replication fork stalling, activate DNA damage responses, and represent hotspots of genomic instability associated with human disease. The 88-bp asymmetric polypurine-polypyrimidine (Pu-Py) mirror repeat tract from the human polycystic kidney disease (PKD1) intron 21 forms non-B DNA secondary structures in vitro. We show that the PKD1 mirror repeat also causes orientation-dependent fork stalling during replication in vitro and in vivo. When integrated alongside the c-myc replicator at an ectopic chromosomal site in the HeLa genome, the Pu-Py mirror repeat tract elicits a polar replication fork barrier. Increased replication protein A (RPA), Rad9, and ataxia telangiectasia- and Rad3-related (ATR) checkpoint protein binding near the mirror repeat sequence suggests that the DNA damage response is activated upon replication fork stalling. Moreover, the proximal c-myc origin of replication was not required to cause orientation-dependent checkpoint activation. Cells expressing the replication fork barrier display constitutive Chk1 phosphorylation and continued growth, i.e. checkpoint adaptation. Excision of the Pu-Py mirror repeat tract abrogates the DNA damage response. Adaptation to Chk1 phosphorylation in cells expressing the replication fork barrier may allow the accumulation of mutations that would otherwise be remediated by the DNA damage response.


Assuntos
Canais de Cátion TRPP/metabolismo , Imunoprecipitação da Cromatina , Citosol/metabolismo , DNA/química , DNA/metabolismo , Dano ao DNA , Primers do DNA/genética , Replicação do DNA , Instabilidade Genômica , Células HeLa , Humanos , Íntrons , Conformação de Ácido Nucleico , Fosforilação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Purinas/química , Pirimidinas/química , Canais de Cátion TRPP/genética
19.
Pediatr Nephrol ; 28(3): 453-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23142864

RESUMO

BACKGROUND: Limited data exist on the performance of cystatin C-based glomerular filtration rate (GFR) equations in pediatric transplant recipients and other high-risk patients. The aim of our study was therefore to evaluate the performance of current cystatin C-based equations in this population. METHODS: This was a retrospective, cross-sectional study of 141 consecutive patients (58 % post-transplant) who received a nuclear medicine GFR (NucGFR) examination using (99m)Tc- diethylenetriaminepentaacetic acid at our institution. Subjects included children receiving liver, kidney or hematopoietic stem cell transplants and patients with oncologic or urologic disease. GFR estimates using published GFR estimating equations, including those based on cystatin C (Filler, Zappitelli, Larsson, Hoek, Rule and Le Bricon equations, respectively) and on both cystatin C and creatinine (Zappitelli, Bouvet and Schwartz equations, respectively), were evaluated and compared to the NucGFR measurement using Bland-Altman analysis. RESULTS: The mean NucGFR was 95 (interquartile range 76-111) ml/min/1.73 m(2). Of the cystatin C-based equations, the Rule, Hoek, Zappitelli and Schwartz (2009 CKiD equation) formulas provided the closest agreement to the NucGFR estimate. All other formulas overestimated the GFR in our cohort. CONCLUSION: Cystatin C-based GFR formulas can provide an accurate estimation of NucGFR in a pediatric population with a high proportion of transplant recipients and oncology patients.


Assuntos
Cistatina C/sangue , Taxa de Filtração Glomerular , Rim/metabolismo , Adolescente , Fatores Etários , Biomarcadores/sangue , Criança , Estudos Transversais , Feminino , Humanos , Rim/diagnóstico por imagem , Rim/fisiopatologia , Nefropatias/sangue , Nefropatias/diagnóstico , Nefropatias/diagnóstico por imagem , Nefropatias/etiologia , Nefropatias/fisiopatologia , Masculino , Modelos Biológicos , Neoplasias/complicações , Transplante de Órgãos/efeitos adversos , Valor Preditivo dos Testes , Cintilografia , Compostos Radiofarmacêuticos , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Pentetato de Tecnécio Tc 99m , Doenças Urológicas/complicações
20.
Nucleic Acids Res ; 39(17): 7465-76, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21653549

RESUMO

The human DEK gene is frequently overexpressed and sometimes amplified in human cancer. Consistent with oncogenic functions, Dek knockout mice are partially resistant to chemically induced papilloma formation. Additionally, DEK knockdown in vitro sensitizes cancer cells to DNA damaging agents and induces cell death via p53-dependent and -independent mechanisms. Here we report that DEK is important for DNA double-strand break repair. DEK depletion in human cancer cell lines and xenografts was sufficient to induce a DNA damage response as assessed by detection of γH2AX and FANCD2. Phosphorylation of H2AX was accompanied by contrasting activation and suppression, respectively, of the ATM and DNA-PK pathways. Similar DNA damage responses were observed in primary Dek knockout mouse embryonic fibroblasts (MEFs), along with increased levels of DNA damage and exaggerated induction of senescence in response to genotoxic stress. Importantly, Dek knockout MEFs exhibited distinct defects in non-homologous end joining (NHEJ) when compared to their wild-type counterparts. Taken together, the data demonstrate new molecular links between DEK and DNA damage response signaling pathways, and suggest that DEK contributes to DNA repair.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas Oncogênicas/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Camundongos , Camundongos Knockout , Camundongos Nus , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas Oncogênicas/genética , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA