Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 314
Filtrar
1.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055068

RESUMO

BACKGROUND: Polycystic kidney disease (PKD) is a genetic disorder affecting millions of people worldwide that is characterized by fluid-filled cysts and leads to end-stage renal disease (ESRD). The hallmarks of PKD are proliferation and dedifferentiation of tubular epithelial cells, cellular processes known to be regulated by Notch signaling. METHODS: We found increased Notch3 expression in human PKD and renal cell carcinoma biopsies. To obtain insight into the underlying mechanisms and the functional consequences of this abnormal expression, we developed a transgenic mouse model with conditional overexpression of the intracellular Notch3 (ICN3) domain specifically in renal tubules. We evaluated the alterations in renal function (creatininemia, BUN) and structure (cysts, fibrosis, inflammation) and measured the expression of several genes involved in Notch signaling and the mechanisms of inflammation, proliferation, dedifferentiation, fibrosis, injury, apoptosis and regeneration. RESULTS: After one month of ICN3 overexpression, kidneys were larger with tubules grossly enlarged in diameter, with cell hypertrophy and hyperplasia, exclusively in the outer stripe of the outer medulla. After three months, mice developed numerous cysts in proximal and distal tubules. The cysts had variable sizes and were lined with a single- or multilayered, flattened, cuboid or columnar epithelium. This resulted in epithelial hyperplasia, which was observed as protrusions into the cystic lumen in some of the renal cysts. The pre-cystic and cystic epithelium showed increased expression of cytoskeletal filaments and markers of epithelial injury and dedifferentiation. Additionally, the epithelium showed increased proliferation with an aberrant orientation of the mitotic spindle. These phenotypic tubular alterations led to progressive interstitial inflammation and fibrosis. CONCLUSIONS: In summary, Notch3 signaling promoted tubular cell proliferation, the alignment of cell division, dedifferentiation and hyperplasia, leading to cystic kidney diseases and pre-neoplastic lesions.


Assuntos
Células Epiteliais/metabolismo , Túbulos Renais/metabolismo , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/metabolismo , Receptor Notch3/metabolismo , Animais , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Células Epiteliais/patologia , Fibrose , Expressão Gênica , Imuno-Histoquímica , Neoplasias Renais/etiologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Túbulos Renais/patologia , Camundongos , Doenças Renais Policísticas/patologia , Receptor Notch3/genética
2.
J Am Soc Nephrol ; 32(9): 2210-2222, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34045314

RESUMO

BACKGROUND: Primary cilia regulation of renal function and BP in health and disease is incompletely understood. This study investigated the effect of nephron ciliary loss on renal physiology, BP, and ensuing cystogenesis. METHODS: Mice underwent doxycycline (DOX)-inducible nephron-specific knockout (KO) of the Ift88 gene at 2 months of age using a Cre-LoxP strategy. BP, kidney function, and renal pathology were studied 2 and 9 months after DOX (Ift88 KO) or vehicle (control). RESULTS: At 2 months post-DOX, male, but not female, Ift88 KO, compared with sex-matched control, mice had reduced BP, enhanced salt-induced natriuresis, increased urinary nitrite and nitrate (NOx) excretion, and increased kidney NOS3 levels, which localized to the outer medulla; the reductions in BP in male mice were prevented by L-NAME. At 9 months post-DOX, male, but not female, Ift88 KO mice had polycystic kidneys, elevated BP, and reduced urinary NOx excretion. No differences were observed in plasma renin concentration, plasma aldosterone, urine vasopressin, or urine PGE2 between Ift88 KO and control mice at 2 or 9 months post-DOX. CONCLUSIONS: Nephron cilia disruption in male, but not female, mice (1) reduces BP prior to cyst formation, (2) increases NOx production that may account for the lower BP prior to cyst formation, and (3) induces polycystic kidneys that are associated with hypertension and reduced renal NO production.


Assuntos
Pressão Sanguínea/fisiologia , Néfrons/fisiopatologia , Doenças Renais Policísticas/etiologia , Proteínas Supressoras de Tumor/genética , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Natriurese , Nitratos/urina , Óxido Nítrico Sintase Tipo III/metabolismo , Nitritos/urina , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Fatores Sexuais
3.
EMBO J ; 40(4): e105120, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33368531

RESUMO

Autophagy is a lysosome-dependent degradation pathway essential to maintain cellular homeostasis. Therefore, either defective or excessive autophagy may be detrimental for cells and tissues. The past decade was characterized by significant advances in molecular dissection of stimulatory autophagy inputs; however, our understanding of the mechanisms that restrain autophagy is far from complete. Here, we describe a negative feedback mechanism that limits autophagosome biogenesis based on the selective autophagy-mediated degradation of ATG13, a component of the ULK1 autophagy initiation complex. We demonstrate that the centrosomal protein OFD1 acts as bona fide autophagy receptor for ATG13 via direct interaction with the Atg8/LC3/GABARAP family of proteins. We also show that patients with Oral-Facial-Digital type I syndrome, caused by mutations in the OFD1 gene, display excessive autophagy and that genetic inhibition of autophagy in a mouse model of the disease, significantly ameliorates polycystic kidney, a clinical manifestation of the disorder. Collectively, our data report the discovery of an autophagy self-regulated mechanism and implicate dysregulated autophagy in the pathogenesis of renal cystic disease in mammals.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagossomos/fisiologia , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Autofagia , Proteínas Associadas aos Microtúbulos/metabolismo , Doenças Renais Policísticas/patologia , Proteínas/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Família da Proteína 8 Relacionada à Autofagia/genética , Humanos , Lisossomos/metabolismo , Lisossomos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/metabolismo , Proteínas/genética
4.
Molecules ; 25(22)2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33238462

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) causes progressive cystic degeneration of the renal tubules, the nephrons, eventually severely compromising kidney function. ADPKD is incurable, with half of the patients eventually needing renal replacement. Treatments for ADPKD patients are limited and new effective therapeutics are needed. Melatonin, a central metabolic regulator conserved across all life kingdoms, exhibits oncostatic and oncoprotective activity and no detected toxicity. Here, we used the Bicaudal C (BicC) Drosophila model of polycystic kidney disease to test the cyst-reducing potential of melatonin. Significant cyst reduction was found in the renal (Malpighian) tubules upon melatonin administration and suggest mechanistic sophistication. Similar to vertebrate PKD, the BicC fly PKD model responds to the antiproliferative drugs rapamycin and mimics of the second mitochondria-derived activator of caspases (Smac). Melatonin appears to be a new cyst-reducing molecule with attractive properties as a potential candidate for PKD treatment.


Assuntos
Cistos/patologia , Melatonina/farmacologia , Doenças Renais Policísticas/patologia , Animais , Animais Geneticamente Modificados , Biomarcadores , Cistos/tratamento farmacológico , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteínas de Drosophila/genética , Feminino , Técnicas de Inativação de Genes , Túbulos Renais/metabolismo , Mutação , Estresse Oxidativo , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/etiologia , Proteínas de Ligação a RNA/genética
6.
EBioMedicine ; 60: 102986, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32949996

RESUMO

BACKGROUND: Polycystic kidney disease (PKD) involves renal cysts arising from proliferating tubular cells. Autophagy has been recently suggested as a potential therapeutic target in PKD, and mammalian target of rapamycin (mTOR) is a key negative regulator of autophagy. However, the effect of autophagy regulation on cystogenesis has not been elucidated in PKD mice. METHODS: Clinical validation was performed using GEO datasets and autosomal dominant polycystic kidney disease (ADPKD) patient samples. Newly established PKD and LC3 transgenic mice were used for in vivo verifications, and additional tests were performed in vitro and in vivo using multiple autophagy drugs. FINDINGS: Neither autophagy stimulation nor LC3 overexpression alleviated PKD. Furthermore, we observed the inhibitory effect of an autophagy inhibitor on cysts, indicating its possible therapeutic use in a specific group of patients with ADPKD. INTERPRETATION: Our findings provide a novel insight into the pathogenesis related to autophagy in PKD, suggesting that drugs related to autophagy regulation should be considered with caution for treating PKD. FUNDING SOURCES: This work was supported by grants from the Bio & Medical Technology Development Program; the Collaborative Genome Program for Fostering New Post-Genome Industry of the NRF; the Basic Science Program.


Assuntos
Autofagia , Cistos/patologia , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/patologia , Animais , Autofagia/genética , Linhagem Celular , Biologia Computacional , Modelos Animais de Doenças , Suscetibilidade a Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Transgênicos , Fosforilação , Doenças Renais Policísticas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Peixe-Zebra
7.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32630605

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common inheritable cause of end stage renal disease and, as of today, only a single moderately effective treatment is available for patients. Even though ADPKD research has made huge progress over the last decades, the precise disease mechanisms remain elusive. However, a wide variety of cellular and animal models have been developed to decipher the pathophysiological mechanisms and related pathways underlying the disease. As none of these models perfectly recapitulates the complexity of the human disease, the aim of this review is to give an overview of the main tools currently available to ADPKD researchers, as well as their main advantages and limitations.


Assuntos
Rim Policístico Autossômico Dominante/etiologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Animais , Cistos/complicações , Modelos Animais de Doenças , Progressão da Doença , Rim/patologia , Falência Renal Crônica/complicações , Modelos Biológicos , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/metabolismo , Suínos , Porco Miniatura
8.
J Vet Diagn Invest ; 32(4): 549-555, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32687010

RESUMO

Polycystic kidney disease (PKD) is one of the most common hereditary diseases in cats, with high prevalence in Persian and Persian-related cats. PKD is caused mainly by an inherited autosomal dominant (AD) mutation, and animals may be asymptomatic for years. We screened 16 cats from various breeds exhibiting a renal abnormality by ultrasound examination and genotyped them for the c.10063C>A transversion on exon 29 of the polycystin-1 (PKD1) gene, by PCR-restriction fragment length polymorphism (PCR-RFLP). Among these cats, a Siamese nuclear family of 4 cats with ancestral hereditary renal failure were screened by whole-genome sequencing (WGS) to determine novel variations in genes associated with both AD and autosomal recessive PKD in humans. During the study period, one cat died as a result of renal failure and was forwarded for autopsy. Additionally, we screened 294 cats asymptomatic for renal disease (Angora, Van, Persian, Siamese, Scottish Fold, Exotic Shorthair, British Shorthair, and mixed breeds) to determine the prevalence of the mutation in cats in Turkey. Ten of the symptomatic and 2 of the asymptomatic cats carried the heterozygous C → A transversion, indicating a prevalence of 62.5% and 0.68%, respectively. In the WGS analysis of 4 cats in the Siamese nuclear family, novel variations were determined in the fibrocystin gene (PKHD1), which was not compatible with dominant inheritance of PKD.


Assuntos
Doenças do Gato/epidemiologia , Mutação , Doenças Renais Policísticas/veterinária , Canais de Cátion TRPP/genética , Animais , Doenças do Gato/etiologia , Doenças do Gato/genética , Gatos , Doenças Renais Policísticas/epidemiologia , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/genética , Reação em Cadeia da Polimerase/veterinária , Polimorfismo de Fragmento de Restrição , Prevalência , Insuficiência Renal/epidemiologia , Insuficiência Renal/etiologia , Insuficiência Renal/genética , Insuficiência Renal/veterinária , Canais de Cátion TRPP/metabolismo , Turquia/epidemiologia , Sequenciamento Completo do Genoma/veterinária
9.
Transplant Proc ; 52(1): 140-145, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31901330

RESUMO

BACKGROUND: Chronic kidney disease (CKD) is a pathophysiological process with many etiologic causes, often leading to end-stage renal disease (ESRD). The distribution of the causes that lead to ESRD varies by country, race, and sex. Renal failure may be prevented by determining these differences and reducing the risk factors. OBJECTIVE: The purpose of the study was to determine the causes and risk factors of previous ESRD in kidney transplant (KT) recipients. METHODS: In this descriptive, cross-sectional study, 393 KT recipients fitting the study criteria gave written consent to participate. Data were collected in face-to-face interviews at the Transplant Center using survey forms prepared by the researchers. RESULTS: According to a multivariate logistic regression analysis of the dependent variable of ESRD diagnosis age of KT recipients, the factors affecting ESRD diagnosis age were found to be job (Odds ratio (OD) = 5.76; 95% CI [2.291-14.481]), diabetes mellitus (DM) (OD = 2.94; 95% CI [1.143-7.571]), polycystic kidney disease (PKD) (OD = 4.55; 95% CI [1.737-11.919]), hypertension (HT) (OD = 3.53; 95% CI [2.132-5.854]), family history of ESRD (OD = 0.57; 95% CI [0.341-0.963]), surgical procedure history (OD = 1.93; 95% CI [1.150-3.230]), and stress level (OD = 5.86, 95% CI [2.212-15.528]). CONCLUSION: It is important that we determine the changeable risk factors related to ESRD development in order to prepare strategies aimed at preventing ESRD, the frequency and prevalence of which is gradually increasing. Modifiable risk factors should be identified, particularly in KT recipients, to preserve the functions of the transplanted kidney.


Assuntos
Falência Renal Crônica/etiologia , Transplante de Rim , Doenças Renais Policísticas/etiologia , Insuficiência Renal Crônica/etiologia , Adulto , Estudos Transversais , Feminino , Humanos , Falência Renal Crônica/epidemiologia , Falência Renal Crônica/cirurgia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Razão de Chances , Doenças Renais Policísticas/epidemiologia , Doenças Renais Policísticas/cirurgia , Prevalência , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/cirurgia , Fatores de Risco
10.
Nat Rev Nephrol ; 15(12): 735-749, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31488901

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) affects an estimated 1 in 1,000 people and slowly progresses to end-stage renal disease (ESRD) in about half of these individuals. Tolvaptan, a vasopressin 2 receptor blocker, has been approved by regulatory authorities in many countries as a therapy to slow cyst growth, but additional treatments that target dysregulated signalling pathways in cystic kidney and liver are needed. Metabolic reprogramming is a prominent feature of cystic cells and a potentially important contributor to the pathophysiology of ADPKD. A number of pathways previously implicated in the pathogenesis of the disease, such as dysregulated mTOR and primary ciliary signalling, have roles in metabolic regulation and may exert their effects through this mechanism. Some of these pathways are amenable to manipulation through dietary modifications or drug therapies. Studies suggest that polycystin-1 and polycystin-2, which are encoded by PKD1 and PKD2, respectively (the genes that are mutated in >99% of patients with ADPKD), may in part affect cellular metabolism through direct effects on mitochondrial function. Mitochondrial dysfunction could alter the redox state and cellular levels of acetyl-CoA, resulting in altered histone acetylation, gene expression, cytoskeletal architecture and response to cellular stress, and in an immunological response that further promotes cyst growth and fibrosis.


Assuntos
Doenças Renais Policísticas/metabolismo , Animais , Humanos , Rim/metabolismo , Rim/fisiopatologia , Redes e Vias Metabólicas , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/patologia
11.
Medicine (Baltimore) ; 98(20): e15600, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31096464

RESUMO

INTRODUCTION: Congenital hepatic fibrosis (CHF) is a rare autosomal recessive disease derived from biliary dysgenesis secondary to ductal plate malformation and is often accompanied by renal cysts or increased renal echogenicity. PATIENT CONCERNS: A 25-year-old woman was admitted to our hospital with splenomegaly and hepatic cirrhosis of a 3-month duration and fever accompanied by abdominal pain for 3 days. The second patient was a 25-year-old male referred to our hospital with hepatomegaly and splenomegaly of 6-year duration who had experienced fever for 3 months and abdominal distension for 1 week. Both 25-year-old patients were found to have CHF with polycystic kidney disease. DIAGNOSIS: Radiological imaging, including computed tomography (CT), magnetic resonance imaging (MRI), and sonography, revealed hepatic fibrosis, portal hypertension, splenomegaly, ascites, bile duct malformation, polycystic kidneys, and CHF. For the first patient, a liver biopsy confirmed the pathological features of CHF, and genetic testing revealed three heterozygous missense mutations, which were classified as "undetermined" in the public Wilson's disease/ATP7B and ADPKD/PKD1 databases. INTERVENTIONS: The first patient had undergone a splenectomy for anemia 2 months previously. Because there is no radical cure for CHF, and due to economic reasons, neither patient received liver transplantation. Therefore, we administered only anti-fibrotic supportive treatment for symptoms. OUTCOMES: Both patients were discharged after their symptoms improved, and both survived for 2 years of follow-up. CONCLUSION: These cases highlight the value of radiological imaging, pathological examination, and genetic evaluation for the diagnosis of CHF. When an individual with unexplained cirrhosis presents with bile duct dilation and malformation as well as polycystic kidneys, the possibility of CHF should be considered. For individuals found to have polycystic kidneys at a young age, the results of liver function tests and imaging examinations including Fibroscan imaging should be continuously and dynamically monitored to enable early diagnosis of CHF.


Assuntos
Doenças Genéticas Inatas/diagnóstico , Cirrose Hepática/diagnóstico , Adulto , Doenças dos Ductos Biliares/etiologia , Feminino , Doenças Genéticas Inatas/complicações , Doenças Genéticas Inatas/diagnóstico por imagem , Doenças Genéticas Inatas/patologia , Humanos , Hipertensão Portal/etiologia , Cirrose Hepática/complicações , Cirrose Hepática/diagnóstico por imagem , Cirrose Hepática/patologia , Masculino , Doenças Renais Policísticas/etiologia , Esplenomegalia/etiologia
12.
J Inherit Metab Dis ; 41(6): 955-963, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29869165

RESUMO

Glycogen storage disease type I (GSDI) is a rare genetic pathology characterized by glucose-6 phosphatase (G6Pase) deficiency, translating in hypoglycemia during short fasts. Besides metabolic perturbations, GSDI patients develop long-term complications, especially chronic kidney disease (CKD). In GSDI patients, CKD is characterized by an accumulation of glycogen and lipids in kidneys, leading to a gradual decline in renal function. At a molecular level, the activation of the renin-angiotensin system is responsible for the development of renal fibrosis, eventually leading to renal failure. The same CKD phenotype was observed in a mouse model with a kidney-specific G6Pase deficiency (K.G6pc-/- mice). Furthermore, GSDI patients and mice develop frequently renal cysts at late stages of the nephropathy, classifying GSDI as a potential polycystic kidney disease (PKD). PKDs are genetic disorders characterized by multiple renal cyst formation, frequently caused by the loss of expression of polycystic kidney genes, such as PKD1/2 and PKHD1. Interestingly, these genes are deregulated in K.G6pc-/- kidneys, suggesting their possible role in GSDI cystogenesis. Finally, renal cysts are known to predispose to renal malignancy development. In addition, HNF1B loss is a malignancy prediction factor. Interestingly, Hnf1b expression was decreased in K.G6pc-/- kidneys. While a single case of renal cancer has been reported in a GSDI patient, a clear cell renal carcinoma was recently observed in one K.G6pc-/- mouse (out of 36 studied mice) at a later stage of the disease. This finding highlights the need to further analyze renal cyst development in GSDI patients in order to evaluate the possible associated risk of carcinogenesis, even if the risk might be limited.


Assuntos
Doença de Depósito de Glicogênio Tipo I/complicações , Doenças Renais Policísticas/etiologia , Insuficiência Renal Crônica/etiologia , Animais , Modelos Animais de Doenças , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/genética , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Neoplasias Renais/etiologia , Neoplasias Renais/patologia , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Insuficiência Renal Crônica/patologia
13.
Nephrol Dial Transplant ; 33(3): 489-496, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28387829

RESUMO

Background: Polycystic kidney disease (PKD) is characterized by urinary tract infections and extrarenal abnormalities such as an increased risk of cancer. As mutations in polycystin-1 and -2 are associated with decreased proliferation of immortalized lymphoblastoid cells in PKD, we investigated whether lymphopenia could be an unrecognized trait of PKD. Methods: We studied 700 kidney transplant recipients with (n = 126) or without PKD at the time of kidney transplantation between 1 January 2003 and 31 December 2014 at Ghent University Hospital. We also studied 204 patients with chronic kidney disease (CKD) with PKD and 204 matched CKD patients without PKD across comparable CKD strata with assessment between 1 January 1999 and 1 February 2016 at three renal outpatient clinics. We compared lymphocyte counts with multiple linear regression analysis to adjust for potential confounders. We analysed flow cytometric immunophenotyping data and other haematological parameters. Results: Lymphocyte counts were 264/µL [95% confidence interval (CI) 144-384] and 345/µL (95% CI 245-445) (both P < 0.001) lower in the end-stage kidney disease (ESKD) and CKD cohort, respectively, after adjustment for age, sex, ln(C-reactive protein) and estimated glomerular filtration rate (in the CKD cohort only). In particular, CD8+ T and B lymphocytes were significantly lower in transplant recipients with versus without PKD (P < 0.001 for both). Thrombocyte and monocyte counts were lower in patients with versus without PKD in both cohorts (P < 0.001 for all analyses except P = 0.01 for monocytes in the ESKD cohort). Conclusion: PKD is characterized by distinct cytopenias and especially lymphopenia, independent of kidney function. This finding has the potential to alter our therapeutic approach to patients with PKD.


Assuntos
Nefropatias/complicações , Linfopenia/complicações , Doenças Renais Policísticas/etiologia , Doenças Renais Policísticas/patologia , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico
14.
Compr Physiol ; 7(3): 945-975, 2017 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-28640449

RESUMO

Renal cysts, which arise from renal tubules, can be seen in a variety of hereditary and nonhereditary entities. Common mechanisms associated with renal cyst formation include increased cell proliferation, epithelial fluid secretion, and extracellular matrix remodeling. Hereditary polycystic kidney disease (PKD) is seen as a component of numerous diseases. Autosomal dominant (AD) PKD is the most common potentially fatal hereditary disease in humans, causes renal failure in approximately 50% of affected individuals, and accounts for approximately 5% of end stage renal disease cases in the United States. ADPKD is caused by mutation in one of two genes-85% of cases are caused by mutation in PKD1 on chromosome 16 and 15% of cases are caused by mutation in PKD2 on chromosome 4. Polycystin-1, encoded by PKD1, is a large protein, has multiple transmembrane spanning domains, has extracellular regions suggesting a role in cell-cell or cell-matrix interactions, has intracellular domains suggesting a role in signal transduction, and can physically interact with Polycystin-2. Polycystin-2 is smaller, has transmembrane domains, can act as a cation channel with calcium permeability, and may be regulated by Polycystin-1. These proteins, and many others associated with cystic kidney disease, localize to primary cilia, which may act as flow sensors in the kidney; cystic kidney diseases have also been termed ciliopathies. An increasing number of intracellular mechanisms, which are abnormally regulated in PKD, have been described and are potential targets for therapy, which is lacking in this common hereditary disease. © 2017 American Physiological Society. Compr Physiol 7:945-975, 2017.


Assuntos
Doenças Renais Policísticas/diagnóstico , Animais , Ensaios Clínicos como Assunto , Humanos , Doenças Renais Policísticas/complicações , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/etiologia , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
15.
16.
PLoS Genet ; 13(4): e1006694, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28406902

RESUMO

Progressive cystic kidney degeneration underlies diverse renal diseases, including the most common cause of kidney failure, autosomal dominant Polycystic Kidney Disease (PKD). Genetic analyses of patients and animal models have identified several key drivers of this disease. The precise molecular and cellular changes underlying cystogenesis remain, however, elusive. Drosophila mutants lacking the translational regulator Bicaudal C (BicC, the fly ortholog of vertebrate BICC1 implicated in renal cystogenesis) exhibited progressive cystic degeneration of the renal tubules (so called "Malpighian" tubules) and reduced renal function. The BicC protein was shown to bind to Drosophila (d-) myc mRNA in tubules. Elevation of d-Myc protein levels was a cause of tubular degeneration in BicC mutants. Activation of the Target of Rapamycin (TOR) kinase pathway, another common feature of PKD, was found in BicC mutant flies. Rapamycin administration substantially reduced the cystic phenotype in flies. We present new mechanistic insight on BicC function and propose that Drosophila may serve as a genetically tractable model for dissecting the evolutionarily-conserved molecular mechanisms of renal cystogenesis.


Assuntos
Proteínas de Drosophila/genética , Doenças Renais Policísticas/etiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Animais Geneticamente Modificados , Cistos , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Regulação da Expressão Gênica , Túbulos de Malpighi/patologia , Mutação , Doenças Renais Policísticas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima
17.
Nefrología (Madr.) ; 37(1): 87-92, ene.-feb. 2017. graf, tab
Artigo em Espanhol | IBECS | ID: ibc-160603

RESUMO

Presentamos el caso de un varón de 32 años, con síndrome de genes contiguos TSC2/PKD1, que le ocasiona esclerosis tuberosa (ET) y poliquistosis renal autosómica dominante simultáneamente. Evolucionó a enfermedad renal terminal y se realizó trasplante renal a los 12 años. Los riñones presentaban angiomiolipomas (AML), que son tumores benignos frecuentes en pacientes con ET. A los 17 años postrasplante, presentó un cuadro de dolor abdominal, anemización y hematoma retroperitoneal. Dicho hematoma se produjo por el sangrado de los AML. Como tratamiento se realizó embolización selectiva. Nuestro paciente podría haberse beneficiado en el momento del trasplante renal del tratamiento con inhibidores de mTOR. Este fármaco actúa como inmunosupresor y reductor tumoral en la ET, al disminuir el riesgo de rotura y hemorragia. En este paciente no se administró porque cuando se trasplantó no se conocía la relación de los inhibidores de mTOR con la ET. Este caso confirma que, a pesar de tratarse de pacientes trasplantados o en diálisis, el riesgo de sangrado por los AML persiste, por lo cual se propone realizar controles periódicos de los riñones propios y valorar la nefrectomía (AU)


We report the case of a 32-year-old male diagnosed with TSC2/PKD1 contiguous gene syndrome, presenting with tuberous sclerosis (TS) and autosomal dominant polycystic kidney disease simultaneously. He progressed to end-stage renal disease and received a kidney transplant at the age of 12. The native kidneys presented angiomyolipomas (AML), which are common benign tumours in patients with TS. Seventeen years after transplantation, he presented with abdominal pain, anaemia and a retroperitoneal haematoma, the latter caused by renal AML bleeding. Selective embolisation was performed. Our patient could have benefited from the administration of mTOR inhibitors at transplant. This therapy is immunosuppressive and reduces the size of benign tumours in TS as well as the risk of rupture and bleeding. This patient did not receive mTOR inhibitors at the time of the transplant because the relationship between mTOR inhibitors and TS was unknown at that time. This case confirms the persistent risk of renal AML bleeding for both transplanted patients and patients on dialysis. As a result, we would recommend routine check-ups of native kidneys and nephrectomy assessment (AU)


Assuntos
Humanos , Masculino , Adulto , Angiomiolipoma/complicações , Neoplasias Renais/complicações , Síndrome WAGR/complicações , Terapia de Substituição Renal , Hemorragia/etiologia , Diálise Renal/métodos , Insuficiência Renal Crônica/terapia , Esclerose Tuberosa/etiologia , Doenças Renais Policísticas/etiologia , Transplante de Rim
18.
Nat Rev Nephrol ; 12(11): 667-677, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27694979

RESUMO

The rate at which autosomal dominant polycystic kidney disease (ADPKD) progresses to end-stage renal disease varies widely and is determined by genetic and non-genetic factors. The ability to determine the prognosis of children and young adults with ADPKD is important for the effective life-long management of the disease and to enable the efficacy of emerging therapies to be determined. Total kidney volume (TKV) reflects the sum volume of hundreds of individual cysts with potentially devastating effects on renal function. The sequential measurement of TKV has been advanced as a dynamic biomarker of disease progression, yet doubt remains among nephrologists and regulatory agencies as to its usefulness. Here, we review the mechanisms that lead to an increase in TKV in ADPKD, and examine the evidence supporting the conclusion that TKV provides a metric of disease progression that can be used to assess the efficacy of potential therapeutic regimens in children and adults with ADPKD. Moreover, we propose that TKV can be used to monitor treatment efficacy in patients with normal levels of renal function, before the pathologic processes of ADPKD cause extensive fibrosis and irreversible loss of functioning renal tissue.


Assuntos
Progressão da Doença , Rim/patologia , Doenças Renais Policísticas/patologia , Humanos , Tamanho do Órgão , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/etiologia , Rim Policístico Autossômico Dominante/patologia , Prognóstico , Resultado do Tratamento
19.
PLoS One ; 11(8): e0161376, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27537361

RESUMO

BACKGROUND: Anthropometric and metabolic risk factors for all-cause end-stage renal disease (ESRD) may vary in their impact depending on the specific primary renal disease. METHODS: In this Austrian population-based prospective cohort study (n = 185,341; 53.9% women) the following data were collected between 1985 and 2005: age, sex, body mass index (BMI), fasting blood glucose (FBG) from 1988, blood pressure, total cholesterol (TC), triglycerides (TG), gamma-glutamyl transferase (GGT) and smoking status. These data were merged with the Austrian Dialysis and Transplant Registry to identify ESRD patients. Cox proportional hazards models were applied to calculate hazard ratios (HR) for all-cause ESRD as well as for cause-specific ESRD due to the following primary renal diseases: autosomal dominant polycystic kidney disease (ADPKD), vascular nephropathy (VN), diabetic nephropathy (DN) and other diseases (OD). RESULTS: During a mean follow-up of 17.5 years 403 participants developed ESRD (ADPKD 36, VN 97, DN 86, and OD 184). All parameters except TG and GGT were significantly associated with all-cause ESRD risk. Particular cause-specific ESRD risk factor patterns were found: for ADPKD increased risk from hypertension (HR 11.55); for VN from smoking (HR 1.81), hypertension (HR 2.37), TG (≥5.70 vs. <1.17 mmol/L: HR 9.27); for DN from smoking (HR 1.77), BMI (≥30 vs. 18.5-24.9 kg/m2: HR 7.55), FBG (≥6.94 vs. <5.55 mmol/L: HR 7.67), hypertension (HR 1.08), TG (≥5.70 vs. <1.17 mmol/L: HR 2.02), GGT (HR 2.14); and for OD from hypertension (HR 2.29), TG (≥5.70 vs. <1.17 mmol/L: HR 6.99) and TC (≥6.22 vs. <5.18 mmol/L: HR 1.56). CONCLUSIONS: Particular anthropometric and metabolic ESRD risk factors differ in importance depending on the primary renal disease. This needs to be considered for future preventive and therapeutic strategies addressing cause-specific ESRD.


Assuntos
Falência Renal Crônica/etiologia , Adulto , Áustria/epidemiologia , Glicemia/análise , Pressão Sanguínea , Índice de Massa Corporal , Colesterol/sangue , Nefropatias Diabéticas/etiologia , Feminino , Humanos , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Masculino , Pessoa de Meia-Idade , Doenças Renais Policísticas/etiologia , Modelos de Riscos Proporcionais , Estudos Prospectivos , Fatores de Risco , Fumar/efeitos adversos , Triglicerídeos/sangue , gama-Glutamiltransferase/sangue
20.
Nat Rev Nephrol ; 12(10): 587-609, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27477490

RESUMO

The mTOR pathway has a central role in the regulation of cell metabolism, growth and proliferation. Studies involving selective gene targeting of mTOR complexes (mTORC1 and mTORC2) in renal cell populations and/or pharmacologic mTOR inhibition have revealed important roles of mTOR in podocyte homeostasis and tubular transport. Important advances have also been made in understanding the role of mTOR in renal injury, polycystic kidney disease and glomerular diseases, including diabetic nephropathy. Novel insights into the roles of mTORC1 and mTORC2 in the regulation of immune cell homeostasis and function are helping to improve understanding of the complex effects of mTOR targeting on immune responses, including those that impact both de novo renal disease and renal allograft outcomes. Extensive experience in clinical renal transplantation has resulted in successful conversion of patients from calcineurin inhibitors to mTOR inhibitors at various times post-transplantation, with excellent long-term graft function. Widespread use of this practice has, however, been limited owing to mTOR-inhibitor- related toxicities. Unique attributes of mTOR inhibitors include reduced rates of squamous cell carcinoma and cytomegalovirus infection compared to other regimens. As understanding of the mechanisms by which mTORC1 and mTORC2 drive the pathogenesis of renal disease progresses, clinical studies of mTOR pathway targeting will enable testing of evolving hypotheses.


Assuntos
Nefropatias/etiologia , Complexos Multiproteicos/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Células Endoteliais/fisiologia , Células Epiteliais/fisiologia , Fibrose/etiologia , Rejeição de Enxerto/etiologia , Humanos , Isquemia/etiologia , Rim/irrigação sanguínea , Rim/citologia , Rim/patologia , Glomérulos Renais , Transplante de Rim , Túbulos Renais/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Podócitos/fisiologia , Doenças Renais Policísticas/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...