Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 227
Filtrar
1.
JCI Insight ; 6(10)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33886508

RESUMO

Flow-activated Na+ and HCO3- transport in kidney proximal tubules (PT) underlies relatively constant fractional reabsorption during changes in glomerular filtration rate (GFR) or glomerulotubular balance (GTB). In view of hypothesized connections of epithelial cilia to flow sensing, we examined flow-activated transport in 3 polycystic kidney disease-related mouse models based on inducible conditional KO of Pkd1, Pkd2, and Kif3a. PTs were harvested from mice after gene inactivation but prior to cyst formation, and flow-mediated PT transport was measured. We confirm that higher flow increased both Na+ and HCO3- absorption in control mice, and we observed that this flow effect was preserved in PTs of Pkd1-/- and Kif3a-/-mice. However, flow activation was absent in Pkd2+/- and Pkd2-/- PT. In heterozygous (Pkd2+/-) mice, a dopamine receptor 1 (DA1) antagonist (SCH23390) restored transport flow sensitivity. When given chronically, this same antagonist reduced renal cyst formation in Pkd2-/-, as evidenced by reduced kidney weight, BUN, and the cystic index, when compared with untreated mice. In contrast, SCH23390 did not prevent cyst formation in Pkd1-/- mice. These results indicate that Pkd2 is necessary for normal GTB and that restoration of flow-activated transport by DA1 antagonist can slow renal cyst formation in Pkd2-/- mice.


Assuntos
Túbulos Renais Proximais/fisiologia , Doenças Renais Policísticas , Animais , Benzazepinas/farmacologia , Modelos Animais de Doenças , Feminino , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
2.
Am J Kidney Dis ; 77(3): 410-419, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33039432

RESUMO

Primary cilia are specialized sensory organelles that protrude from the apical surface of most cell types. During the past 2 decades, they have been found to play important roles in tissue development and signal transduction, with mutations in ciliary-associated proteins resulting in a group of diseases collectively known as ciliopathies. Many of these mutations manifest as renal ciliopathies, characterized by kidney dysfunction resulting from aberrant cilia or ciliary functions. This group of overlapping and genetically heterogeneous diseases includes polycystic kidney disease, nephronophthisis, and Bardet-Biedl syndrome as the main focus of this review. Renal ciliopathies are characterized by the presence of kidney cysts that develop due to uncontrolled epithelial cell proliferation, growth, and polarity, downstream of dysregulated ciliary-dependent signaling. Due to cystic-associated kidney injury and systemic inflammation, cases result in kidney failure requiring dialysis and transplantation. Of the handful of pharmacologic treatments available, none are curative. It is important to determine the molecular mechanisms that underlie the involvement of the primary cilium in cyst initiation, expansion, and progression for the development of novel and efficacious treatments. This review updates research progress in defining key genes and molecules central to ciliogenesis and renal ciliopathies.


Assuntos
Síndrome de Bardet-Biedl/genética , Cílios/metabolismo , Ciliopatias/genética , Doenças Renais Policísticas/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Síndrome de Bardet-Biedl/metabolismo , Síndrome de Bardet-Biedl/fisiopatologia , Cerebelo/anormalidades , Cerebelo/metabolismo , Cerebelo/fisiopatologia , Chaperoninas/genética , Cílios/fisiologia , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/fisiopatologia , Ciliopatias/metabolismo , Ciliopatias/fisiopatologia , Proteínas do Citoesqueleto/genética , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/fisiopatologia , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/fisiopatologia , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/fisiopatologia , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/metabolismo , Amaurose Congênita de Leber/fisiopatologia , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Atrofias Ópticas Hereditárias/genética , Atrofias Ópticas Hereditárias/metabolismo , Atrofias Ópticas Hereditárias/fisiopatologia , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/fisiopatologia , Proteínas/genética , Retina/anormalidades , Retina/metabolismo , Retina/fisiopatologia , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Retinose Pigmentar/fisiopatologia , Canais de Cátion TRPP/genética
3.
Adv Drug Deliv Rev ; 161-162: 176-189, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32866560

RESUMO

Polycystic kidney disease (PKD) is characterized by progressive cyst growth and is a leading cause of renal failure worldwide. Currently, there are limited therapeutic options available to PKD patients, and only one drug, tolvaptan, has been FDA-approved to slow cyst progression. Similar to other small molecule drugs, however, tolvaptan is costly, only moderately effective, and causes adverse events leading to high patient dropout rates. Peptides may mitigate many drawbacks of small molecule drugs, as they can be highly tissue-specific, biocompatible, and economically scaled-up. Peptides can function as targeting ligands that direct therapies to diseased renal tissue, or be potent as therapeutic agents themselves. This review discusses various aberrant signaling pathways in PKD and renal receptors that can be potential targets of peptide-mediated strategies. Additionally, peptides utilized in other kidney applications, but may prove useful in the context of PKD, are highlighted. Insights into novel peptide-based solutions that have potential to improve clinical management of PKD are provided.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Rim/fisiologia , Peptídeos/administração & dosagem , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/fisiopatologia , Animais , Progressão da Doença , Humanos , Transdução de Sinais
4.
J Am Soc Nephrol ; 31(6): 1178-1190, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32295825

RESUMO

The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.


Assuntos
Nefropatias/etiologia , Vasos Linfáticos/fisiologia , Imunidade Adaptativa , Rejeição de Enxerto , Humanos , Nefropatias/fisiopatologia , Transplante de Rim/efeitos adversos , Linfa/fisiologia , Linfangiogênese , Vasos Linfáticos/anatomia & histologia , Vasos Linfáticos/citologia , Doenças Renais Policísticas/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
5.
Expert Opin Ther Targets ; 24(6): 589-599, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32250187

RESUMO

Introduction: Polycystic liver disease (PLD) is a rare disease defined by the growth of hepatic cysts and occurs either isolated or as an extrarenal manifestation of polycystic kidney disease. While surgery has been the mainstay in treatment of symptomatic PLD, recently discovered regulatory mechanisms affecting hepatic cystogenesis provide potential new therapies to reduce hepatic cyst burden.Areas covered: This review summarizes intracellular pathways and therapeutic targets involved in hepatic cystogenesis. While drugs that target cAMP, mTOR and bile acids were evaluated in clinical trials, investigation in autophagy, Wnt and miRNA signaling pathways are still in the pre-clinical phase. Recent epidemiological data present female hormones as a promising therapeutic target. Additionally, therapeutic advances in renal cystogenesis are reviewed for their potential application in treatment of hepatic cysts.Expert opinion: Further elucidation of the pathophysiology of hepatic cystogenesis is needed to provide additional targets and improve the efficacy of current treatments. The most promising therapeutic target in PLD is the female hormone pathway, given the increased severity in women and the harmful effects of exogenous estrogens. In addition, combining current pharmaceutical and surgical therapies can lead to improved outcomes. Lastly, the rarity of PLD creates the need to share expertise internationally.


Assuntos
Cistos/tratamento farmacológico , Hepatopatias/tratamento farmacológico , Terapia de Alvo Molecular , Doenças Renais Policísticas/tratamento farmacológico , Animais , Autofagia/efeitos dos fármacos , Cistos/fisiopatologia , Feminino , Humanos , Hepatopatias/fisiopatologia , MicroRNAs/metabolismo , Doenças Renais Policísticas/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
6.
Am J Physiol Renal Physiol ; 318(3): F557-F564, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31928223

RESUMO

Klotho interacts with various membrane proteins such as receptors for transforming growth factor-ß (TGF-ß) and insulin-like growth factor (IGF). Renal expression of klotho is diminished in polycystic kidney disease (PKD). In the present study, the effects of klotho supplementation on PKD were assessed. Recombinant human klotho protein (10 µg·kg-1·day-1) or a vehicle was administered daily by subcutaneous injection to 6-wk-old mice with PKD (DBA/2-pcy). Blood pressure was measured using tail-cuff methods. After 2 mo, mice were killed, and the kidneys were harvested for analysis. Exogenous klotho protein supplementation reduced kidney weight, cystic area, systolic blood pressure, renal angiotensin II levels, and 8-epi-PGF2α excretion (P < 0.05). Klotho protein supplementation enhanced glomerular filtration rate, renal expression of superoxide dismutase, and klotho itself (P < 0.05). Klotho supplementation attenuated renal expressions of TGF-ß and collagen type I and diminished renal abundance of Twist, phosphorylated Akt, and mammalian target of rapamycin (P < 0.05). Pathological examination revealed that klotho decreased the fibrosis index and nuclear staining of Smad in PKD kidneys (P < 0.05). Our data indicate that klotho protein supplementation ameliorates the renin-angiotensin system, reducing blood pressure in PKD mice. Furthermore, the present results implicate klotho supplementation in the suppression of Akt/mammalian target of rapamycin signaling, slowing cystic expansion. Finally, our findings suggest that klotho protein supplementation attenuated fibrosis at least partly by inhibiting epithelial mesenchymal transition in PKD.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Glucuronidase/uso terapêutico , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/genética , Animais , Células Cultivadas , Feminino , Glucuronidase/administração & dosagem , Injeções Subcutâneas , Rim/fisiologia , Proteínas Klotho , Camundongos , Miofibroblastos/efeitos dos fármacos , Doenças Renais Policísticas/fisiopatologia , Proteínas Recombinantes
7.
Hum Pathol ; 97: 1-7, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31857138

RESUMO

Nonfunctioning kidneys secondary to various etiologies display different histopathological features. Studies focused on incidence and types of renal neoplasms using the new World Health Organization and International Society of Urological Pathology classification system in various types of nonfunctioning kidneys are very limited. We identified 311 nephrectomies of nonfunctioning kidneys and categorized them into 5 categories: acquired cystic kidney disease (ACKD, n = 61); end-stage renal disease, nonspecific (ESRD, n = 63); adult polycystic kidney disease (APKD, n = 49); failed transplant kidney (FTK, n = 96); and those caused by obstructive conditions in the kidney (OCK, n = 42). ACKD (70%) and ESRD (43%) had higher cancer incidences than the other 3 groups (APKD = 2%, FTK = 0%, and OCK = 5%). Besides clear cell renal cell carcinoma (RCC) and papillary RCC, clear cell papillary RCC had a much higher incidence within ACKD patients (13/61) compared to other groups. ACKD-associated RCC was only identified in ACKD patients. ACKD patients had significantly longer dialysis duration compared to ESRD, APKD, and FTK. Although they had similar risk for clear cell RCC and papillary RCC, ACKD patients had a much higher risk for ACKD-associated RCC and clear cell papillary RCC than ESRD patients. Although most RCCs arising in these nonfunctioning kidneys were early pT1 stage, 6 ACKD patients and 3 ESRD patients had higher-stage diseases, which can be fatal if not treated appropriately. Therefore, precise clinicopathological classification of these nonfunctioning kidneys is important for predicting kidney cancer risk. These results indicate the need for active monitoring of the patients with high-risk nonfunctioning kidney diseases and appropriate surgical treatment when necessary.


Assuntos
Carcinoma de Células Renais/patologia , Doenças Renais Císticas/patologia , Falência Renal Crônica/patologia , Neoplasias Renais/patologia , Doenças Renais Policísticas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/epidemiologia , Carcinoma de Células Renais/fisiopatologia , Carcinoma de Células Renais/cirurgia , Feminino , Humanos , Incidência , Doenças Renais Císticas/epidemiologia , Doenças Renais Císticas/fisiopatologia , Doenças Renais Císticas/terapia , Falência Renal Crônica/epidemiologia , Falência Renal Crônica/fisiopatologia , Falência Renal Crônica/terapia , Neoplasias Renais/epidemiologia , Neoplasias Renais/fisiopatologia , Neoplasias Renais/cirurgia , Transplante de Rim , Masculino , Pessoa de Meia-Idade , Nefrectomia , Doenças Renais Policísticas/epidemiologia , Doenças Renais Policísticas/fisiopatologia , Doenças Renais Policísticas/terapia , Prognóstico , Diálise Renal , Medição de Risco , Fatores de Risco , Falha de Tratamento , Adulto Jovem
8.
J Mol Cell Biol ; 12(8): 644-653, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31065693

RESUMO

Polycystic kidney disease (PKD) is a prevalent genetic disorder, characterized by the formation of kidney cysts that progressively lead to kidney failure. The currently available drug tolvaptan is not well tolerated by all patients and there remains a strong need for alternative treatments. The signaling rewiring in PKD that drives cyst formation is highly complex and not fully understood. As a consequence, the effects of drugs are sometimes difficult to predict. We previously established a high throughput microscopy phenotypic screening method for quantitative assessment of renal cyst growth. Here, we applied this 3D cyst growth phenotypic assay and screened 2320 small drug-like molecules, including approved drugs. We identified 81 active molecules that inhibit cyst growth. Multi-parametric phenotypic profiling of the effects on 3D cultured cysts discriminated molecules that showed preferred pharmacological effects above genuine toxicological properties. Celastrol, a triterpenoid from Tripterygium Wilfordii, was identified as a potent inhibitor of cyst growth in vitro. In an in vivo iKspCre-Pkd1lox,lox mouse model for PKD, celastrol inhibited the growth of renal cysts and maintained kidney function.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Triterpenos Pentacíclicos/uso terapêutico , Doenças Renais Policísticas/tratamento farmacológico , Animais , Cistos/patologia , Cistos/fisiopatologia , Testes de Função Renal , Camundongos , Triterpenos Pentacíclicos/farmacologia , Fenótipo , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico
9.
Adv Exp Med Biol ; 1165: 81-100, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31399962

RESUMO

Polycystic kidney disease (PKD) is a common genetic disorder characterized by formations of numerous cysts in kidneys and most caused by PKD1 or PKD2 mutations in autosomal dominant polycystic kidney disease (ADPKD). The interstitial inflammation and fibrosis is one of the major pathological changes in polycystic kidney tissues with an accumulation of inflammatory cells, chemokines, and cytokines. The immune response is observed across different stages and occurs prior to or coincident with cyst formation in ADPKD. Evidence for inflammation as an important contributor to cyst growth and fibrosis includes increased interstitial macrophages, upregulated expressions of pro-inflammatory cytokines, activated complement system, and activated pathways including NF-κB and JAK-STAT signaling in polycystic kidney tissues. Inflammatory cells are responsible for overproduction of several pro-fibrotic growth factors which promote renal fibrosis in ADPKD. These growth factors trigger epithelial mesenchymal transition and myofibroblast/fibrocyte activation, which stimulate the expansion of extracellular matrix (ECM) including collagen I, III, IV, V, and fibronectin, leading to renal fibrosis and reduced renal function. Besides, there are imbalanced ECM turnover regulators which lead to the increased ECM production and inadequate degradation in polycystic kidney tissues. Several fibrosis associated signaling pathways, such as TGFß-SMAD, Wnt, and periostin-integrin-linked kinase are also activated in polycystic kidney tissues. Although the effective anti-fibrotic treatments are limited at the present time, slowing the cyst expansion and fibrosis development is very important for prolonging life span and improving the palliative care of ADPKD patients. The inhibition of pro-fibrotic cytokines involved in fibrosis might be a new therapeutic strategy for ADPKD in the future.


Assuntos
Rim/patologia , Doenças Renais Policísticas/fisiopatologia , Proteínas do Sistema Complemento , Citocinas , Matriz Extracelular , Fibrose , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Macrófagos , Rim Policístico Autossômico Dominante , Transdução de Sinais
10.
Adv Exp Med Biol ; 1132: 99-112, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037629

RESUMO

Periostin is a matricellular protein that is expressed in several tissues during embryonic development; however, its expression in adults is mostly restricted to collagen-rich connective tissues. Periostin is expressed only briefly during kidney development, but it is not normally detected in the adult kidney. Recent evidence has revealed that periostin is aberrantly expressed in several forms of chronic kidney disease (CKD), and that its expression correlates with the degree of interstitial fibrosis and the decline in renal function. Polycystic kidney disease (PKD), a genetic disorder, is characterized by the formation of numerous fluid-filled cysts in the kidneys. Periostin is secreted by the cyst epithelial cells and accumulates within the extracellular matrix adjacent to the cysts. In PKD mice, periostin overexpression accelerates cyst growth and contributes to structural changes in the kidneys, including interstitial fibrosis. Recent evidence suggests that periostin is a tissue repair molecule; however, its role in repair following acute kidney injury has not been investigated. It is thought that persistent expression of this protein in CKD contributes importantly to tubulointerstitial fibrosis and the progressive decline in renal function. Future studies to define the diverse actions of periostin during kidney injury may lead to effective therapies to slow PKD progression and possibly prevent the development of CKD. This chapter reviews the current literature on the expression of periostin in PKD and other forms of CKD, mechanisms for periostin stimulated cyst growth, its potential role in extracellular matrix production and renal fibrosis, and the evidence for periostin as a novel biomarker for kidney disease.


Assuntos
Moléculas de Adesão Celular/fisiologia , Rim/fisiologia , Insuficiência Renal Crônica/fisiopatologia , Animais , Matriz Extracelular , Fibrose , Humanos , Rim/patologia , Camundongos , Doenças Renais Policísticas/fisiopatologia
11.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31091126

RESUMO

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Assuntos
Arritmias Cardíacas/metabolismo , Cardiomegalia/metabolismo , Quimiocina CCL2/metabolismo , Rim/metabolismo , Pulmão/metabolismo , Miocárdio/metabolismo , Doenças Renais Policísticas/metabolismo , Edema Pulmonar/metabolismo , Animais , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Células Cultivadas , Quimiocina CCL2/deficiência , Quimiocina CCL2/genética , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Rim/patologia , Rim/fisiopatologia , Pulmão/patologia , Pulmão/fisiopatologia , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/patologia , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Edema Pulmonar/patologia , Edema Pulmonar/fisiopatologia , Edema Pulmonar/prevenção & controle , Fatores de Tempo
12.
Pediatr Nephrol ; 34(6): 1065-1075, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30666461

RESUMO

BACKGROUND: HNF1B gene mutations are an important cause of bilateral (cystic) dysplasia in children, complicated by chronic renal insufficiency. The clinical variability, the absence of genotype-phenotype correlations, and limited long-term data render counseling of affected families difficult. METHODS: Longitudinal data of 62 children probands with genetically proven HNF1B nephropathy was obtained in a multicenter approach. Genetic family cascade screening was performed in 30/62 cases. RESULTS: Eighty-seven percent of patients had bilateral dysplasia, 74% visible bilateral, and 16% unilateral renal cysts at the end of observation. Cyst development was non-progressive in 72% with a mean glomerular filtration rate (GFR) loss of - 0.33 ml/min/1.73m2 per year (± 8.9). In patients with an increase in cyst number, the annual GFR reduction was - 2.8 ml/min/1.73m2 (± 13.2), in the total cohort - 1.0 ml/min/1.73m2 (±10.3). A subset of HNF1B patients differs from this group and develops end stage renal disease (ESRD) at very early ages < 2 years. Hyperuricemia (37%) was a frequent finding at young age (median 1 year), whereas hypomagnesemia (24%), elevated liver enzymes (21%), and hyperglycemia (8%) showed an increased incidence in the teenaged child. Genetic analysis revealed no genotype-phenotype correlations but a significant parent-of-origin effect with a preponderance of 81% of maternal inheritance in dominant cases. CONCLUSIONS: In most children, HNF1B nephropathy has a non-progressive course of cyst development and a slow-progressive course of kidney function. A subgroup of patients developed ESRD at very young age < 2 years requiring special medical attention. The parent-of-origin effect suggests an influence of epigenetic modifiers in HNF1B disease.


Assuntos
Fator 1-beta Nuclear de Hepatócito/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Adolescente , Idade de Início , Criança , Pré-Escolar , Progressão da Doença , Feminino , Estudos de Associação Genética , Alemanha , Humanos , Lactente , Recém-Nascido , Falência Renal Crônica/genética , Masculino , Fenótipo , Sistema de Registros
13.
Clin Exp Nephrol ; 23(4): 455-464, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30426292

RESUMO

BACKGROUND: Tolvaptan is an effective treatment for polycystic kidney disease (PKD), but also causes unfortunate polyuria. Hydrochlorothiazide (HCTZ) has been shown to reduce urine volume in nephrogenic diabetes insipidus, raising the possibility that HCTZ could also be effective in reducing tolvaptan-induced polyuria. In this study, we examined the combined administration of HCTZ and tolvaptan. METHODS: Male PCK rats were divided into four groups of normal chow (Cont), normal chow plus tolvaptan, gavage HCTZ treatment, and tolvaptan + HCTZ. Biochemical examinations of the plasma and urine were performed as well as histological and molecular (mRNA and protein expression) analyses. RESULTS: Groups treated with tolvaptan had significantly higher 24 h urine excretion, which was significantly reduced in the tolvaptan + HCTZ group after 2 weeks. Cyst size, pERK protein expression, and Cyclin D1 mRNA expression were all significantly reduced in both the tolvaptan and tolvaptan + HCTZ groups, indicating that HCTZ did not affect the beneficial functions of tolvaptan. Notably, aquaporin 2 redistribution from the apical to intracellular domains was observed in tolvaptan-treated rats and was partially reversed in the tolvaptan + HCTZ group. The renal glomerular filtration rate was reduced in the tolvaptan + HCTZ group. Significantly lowered mRNA expression of neuronal nitric oxide synthase, prostaglandin E synthase 2 and renin were also found in the medulla, but not in the cortex. CONCLUSION: HCTZ reduces tolvaptan-induced polyuria without altering its beneficial effects on PKD. This novel therapeutic combination could potentially lead to better PKD treatments and improved quality of life for the affected patients.


Assuntos
Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Diuréticos/uso terapêutico , Hidroclorotiazida/uso terapêutico , Doenças Renais Policísticas/tratamento farmacológico , Poliúria/tratamento farmacológico , Tolvaptan/uso terapêutico , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/efeitos adversos , Aquaporina 2/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Quimioterapia Combinada , Expressão Gênica/efeitos dos fármacos , Taxa de Filtração Glomerular , Masculino , Óxido Nítrico Sintase Tipo I/genética , Doenças Renais Policísticas/fisiopatologia , Poliúria/induzido quimicamente , Prostaglandina-E Sintases/genética , RNA Mensageiro/metabolismo , Ratos , Renina/genética , Tolvaptan/efeitos adversos , Urina
14.
J Physiol ; 597(2): 499-519, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30242840

RESUMO

KEY POINTS: Significant and selective up-regulation of the Na+ /H+ exchanger NHA2 (SLC9B2) was observed in cysts of patients with autosomal dominant polycystic kidney disease. Using the MDCK cell model of cystogenesis, it was found that NHA2 increases cyst size. Silencing or pharmacological inhibition of NHA2 inhibits cyst formation in vitro. Polycystin-1 represses NHA2 expression via Ca2+ /NFAT signalling whereas the dominant negative membrane-anchored C-terminal fragment (PC1-MAT) increased NHA2 levels. Drugs (caffeine, theophylline) and hormones (vasopressin, aldosterone) known to exacerbate cysts elicit NHA2 expression. Taken together, the findings reveal NHA2 as a potential new player in salt and water homeostasis in the kidney and in the pathogenesis of polycystic kidney disease. ABSTRACT: Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2 encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively. The molecular pathways linking polycystins to cyst development in ADPKD are still unclear. Intracystic fluid secretion via ion transporters and channels plays a crucial role in cyst expansion in ADPKD. Unexpectedly, we observed significant and selective up-regulation of NHA2, a member of the SLC9B family of Na+ /H+ exchangers, that correlated with cyst size and disease severity in ADPKD patients. Using three-dimensional cultures of MDCK cells to model cystogenesis in vitro, we showed that ectopic expression of NHA2 is causal to increased cyst size. Induction of PC1 in MDCK cells inhibited NHA2 expression with concordant inhibition of Ca2+ influx through store-dependent and -independent pathways, whereas reciprocal activation of Ca2+ influx by the dominant negative membrane-anchored C-terminal tail fragment of PC1 elevated NHA2. We showed that NHA2 is a target of Ca2+ /NFAT signalling and is transcriptionally induced by methylxanthine drugs such as caffeine and theophylline, which are contraindicated in ADPKD patients. Finally, we observed robust induction of NHA2 by vasopressin, which is physiologically consistent with increased levels of circulating vasopressin and up-regulation of vasopressin V2 receptors in ADPKD. Our findings have mechanistic implications on the emerging use of vasopressin V2 receptor antagonists such as tolvaptan as safe and effective therapy for polycystic kidney disease and reveal a potential new regulator of transepithelial salt and water transport in the kidney.


Assuntos
Antiporters/genética , Doenças Renais Policísticas , Animais , Antiporters/metabolismo , Técnicas de Cultura de Células , Cistos , Cães , Células HEK293 , Homeostase , Humanos , Rim/metabolismo , Rim/fisiopatologia , Células Madin Darby de Rim Canino , Modelos Biológicos , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/fisiopatologia
15.
Nephron ; 141(1): 50-60, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30359986

RESUMO

Early and severe forms of polycystic kidney disease (PKD) do already manifest during childhood or adolescence. They are characterized by enlarged kidneys and diminished renal function that prenatally may result in Potter's oligohydramnios sequence. Genetically, various defects can mimic this phenotype. Most common are PKHD1 mutations that lead to autosomal recessive PKD (ARPKD). About the same number of children do carry mutations in the dominant autosomal dominant polycystic kidney disease (ADPKD) genes, PKD1 and less frequent PKD2, often arise de novo or may affect both disease alleles in a recessive mode. Mutations in DZIP1L have been recently described to result in an ARPKD-like phenotype. Likewise, mutations in several other cystogenes can phenocopy early and severe PKD. Early and reliable prenatal diagnosis for which there is a strong demand in ARPKD and related diseases is feasible only by genetics. A comprehensive knowledge of disease-causing genes is essential for the correct diagnosis and parental counselling. The increasing number of genes that need to be considered benefits from the advances of next generation sequencing and allows the simultaneous analysis of all genes of interest in a single test, which is now the mainstay for genetic diagnosis. Interpretation of data is challenging and requires expert knowledge in data handling, bioinformatics and clinical genetics.


Assuntos
Ciliopatias/terapia , Doenças Renais Policísticas/terapia , Proteínas Adaptadoras de Transdução de Sinal/genética , Ciliopatias/genética , Ciliopatias/fisiopatologia , Testes Genéticos , Humanos , Mutação/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética
16.
Ann Nutr Metab ; 72 Suppl 2: 33-38, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29925070

RESUMO

Polycystic kidney disease (PKD) is a group of monogenetic conditions characterised by the progressive accumulation of multiple renal cysts and hypertension. One of the earliest features of PKD is a reduction in urinary concentrating capacity that impairs extracellular fluid conservation. Urinary concentrating impairment predisposes PKD patients to periods of hypohydration when fluid loss is not adequately compensated by fluid intake. The hypohydrated state provides a blood hyperosmotic stimulus for vasopressin release to minimise further water loss. However, over-activation of renal V2 receptors contributes to cyst expansion. Although suppressing vasopressin release with high water intake has been shown to impair disease progression in rodent models, whether this approach is efficacious in patients remains uncertain. The neural osmoregulatory pathway that controls vasopressin secretion also exerts a stimulatory action on vasomotor sympathetic activity and blood pressure during dehydration. Recurrent dehydration leads to a worsening of hypertension in rodents and cross-sectional data suggests that reduced urinary concentrating ability may contribute to hypertension development in the clinical PKD population. Experimental studies are required to evaluate this hypothesis and to determine the underlying mechanism.


Assuntos
Hipertensão/fisiopatologia , Osmorregulação , Doenças Renais Policísticas/fisiopatologia , Animais , Progressão da Doença , Ingestão de Líquidos , Humanos , Hipertensão/complicações , Doenças Renais Policísticas/complicações , Receptores de Vasopressinas/fisiologia , Urina/química , Vasopressinas/fisiologia
17.
Genes Dev ; 32(11-12): 781-793, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29891559

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is an inherited disorder caused by mutations in PKD1 or PKD2 and affects one in 500-1000 humans. Limited treatment is currently available for ADPKD. Here we identify the Hippo signaling effector YAP and its transcriptional target, c-Myc, as promoters of cystic kidney pathogenesis. While transgenic overexpression of YAP promotes proliferation and tubule dilation in mouse kidneys, loss of YAP/TAZ or c-Myc suppresses cystogenesis in a mouse ADPKD model resulting from Pkd1 deficiency. Through a comprehensive kinase inhibitor screen based on a novel three-dimensional (3D) culture of Pkd1 mutant mouse kidney cells, we identified a signaling pathway involving the RhoGEF (guanine nucleotide exchange factor) LARG, the small GTPase RhoA, and the RhoA effector Rho-associated kinase (ROCK) as a critical signaling module between PKD1 and YAP. Further corroborating its physiological importance, inhibition of RhoA signaling suppresses cystogenesis in 3D culture of Pkd1 mutant kidney cells as well as Pkd1 mutant mouse kidneys in vivo. Taken together, our findings implicate the RhoA-YAP-c-Myc signaling axis as a critical mediator and potential drug target in ADPKD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Rim/fisiopatologia , Fosfoproteínas/metabolismo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Células HEK293 , Humanos , Rim/citologia , Rim/patologia , Camundongos , Fosfoproteínas/genética , Doenças Renais Policísticas/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas de Sinalização YAP , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP
18.
Brain Dev ; 40(4): 259-267, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29217415

RESUMO

OBJECTIVE: Arima syndrome (AS) is a rare disease and its clinical features mimic those of Joubert syndrome or Joubert syndrome-related diseases (JSRD). Recently, we clarified the AS diagnostic criteria and its severe phenotype. However, genetic evidence of AS remains unknown. We explored causative genes of AS and compared the clinical and genetic features of AS with the other JSRD. PATIENTS AND METHODS: We performed genetic analyses of 4 AS patients of 3 families with combination of whole-exome sequencing and Sanger sequencing. Furthermore, we studied cell biology with the cultured fibroblasts of 3 AS patients. RESULTS: All patients had a specific homozygous variant (c.6012-12T>A, p.Arg2004Serfs*7) or compound heterozygous variants (c.1711+1G>A; c.6012-12T>A, p.Gly570Aspfs*19;Arg2004Serfs*7) in centrosomal protein 290 kDa (CEP290) gene. These unique variants lead to abnormal splicing and premature termination. Morphological analysis of cultured fibroblasts from AS patients revealed a marked decrease of the CEP290-positive cell number with significantly longer cilium and naked and protruded ciliary axoneme without ciliary membrane into the cytoplasm. CONCLUSION: AS resulted in cilia dysfunction from centrosome disruption. The unique variant of CEP290 could be strongly linked to AS pathology. Here, we provided AS specific genetic evidence, which steers the structure and functions of centrosome that is responsible for normal ciliogenesis. This is the first report that has demonstrated the molecular basis of Arima syndrome.


Assuntos
Antígenos de Neoplasias/genética , Doenças Cerebelares/genética , Doenças Cerebelares/patologia , Coloboma/genética , Coloboma/patologia , Fibroblastos/patologia , Proteínas de Neoplasias/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Anormalidades Múltiplas/patologia , Anormalidades Múltiplas/fisiopatologia , Adolescente , Adulto , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Centrossomo/metabolismo , Centrossomo/patologia , Doenças Cerebelares/fisiopatologia , Cerebelo/anormalidades , Cerebelo/patologia , Cerebelo/fisiopatologia , Cílios/metabolismo , Cílios/patologia , Coloboma/fisiopatologia , Proteínas do Citoesqueleto , Anormalidades do Olho/patologia , Anormalidades do Olho/fisiopatologia , Família , Feminino , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Doenças Renais Císticas/patologia , Doenças Renais Císticas/fisiopatologia , Microscopia Eletrônica de Transmissão , Peso Molecular , Mutação , Proteínas de Neoplasias/metabolismo , Doenças Renais Policísticas/fisiopatologia , Retina/anormalidades , Retina/patologia , Retina/fisiopatologia , Sequenciamento do Exoma , Adulto Jovem
19.
Int Ophthalmol ; 38(4): 1559-1564, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28664236

RESUMO

PURPOSE: The aim of this study was to evaluate the corneal biomechanical features in polycystic kidney disease (PKD) patients and compare them with the healthy individuals. METHODS: Totally 81 patients with a mean age of 48.46 ± 14.51 years and 60 control cases with a mean age of 44.68 ± 12.69 years were included in the study. All of the subjects underwent a complete ophthalmological examination, including visual acuity testing, biomicroscopic anterior and posterior segment examinations. Corneal hysteresis (CH), corneal resistance factor (CRF), Goldmann-correlated intraocular pressure (IOPg) and corneal-compensated intraocular pressure (IOPcc) were evaluated with the ocular response analyzer, and the central corneal thickness was evaluated with Sirius® corneal topography. RESULTS: PKD patients had significantly increased CH values, without any alterations in IOP or CCT values, compared with the control cases (p:0.001). Among PKD patients, 23 were having liver cysts accompanying renal cysts. There was not any statistically significant difference between PKD patients with or without liver cysts regarding biomechanical properties of the cornea. However, both patient groups had statistically significantly increased CH values compared with the control cases. CONCLUSION: Patients with PKD present with higher CH values than age-matched controls. Larger studies are warranted to elucidate the alterations in corneal biomechanical properties and their clinical relevance in PKD patients.


Assuntos
Córnea/fisiologia , Elasticidade/fisiologia , Doenças Renais Policísticas/fisiopatologia , Adulto , Idoso , Fenômenos Biomecânicos , Estudos de Casos e Controles , Humanos , Pressão Intraocular/fisiologia , Pessoa de Meia-Idade
20.
Curr Biol ; 27(20): 3120-3131.e4, 2017 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-29033332

RESUMO

Oriented cell division (OCD) and convergent extension (CE) shape developing renal tubules, and their disruption has been associated with polycystic kidney disease (PKD) genes, the majority of which encode proteins that localize to primary cilia. Core planar cell polarity (PCP) signaling controls OCD and CE in other contexts, leading to the hypothesis that disruption of PCP signaling interferes with CE and/or OCD to produce PKD. Nonetheless, the contribution of PCP to tubulogenesis and cystogenesis is uncertain, and two major questions remain unanswered. Specifically, the inference that mutation of PKD genes interferes with PCP signaling is untested, and the importance of PCP signaling for cystogenic PKD phenotypes has not been examined. We show that, during proliferative stages, PCP signaling polarizes renal tubules to control OCD. However, we find that, contrary to the prevailing model, PKD mutations do not disrupt PCP signaling but instead act independently and in parallel with PCP signaling to affect OCD. Indeed, PCP signaling that is normally downregulated once development is completed is retained in cystic adult kidneys. Disrupting PCP signaling results in inaccurate control of tubule diameter, a tightly regulated parameter with important physiological ramifications. However, we show that disruption of PCP signaling is not cystogenic. Our results suggest that regulating tubule diameter is a key function of PCP signaling but that loss of this control does not induce cysts.


Assuntos
Polaridade Celular/fisiologia , Túbulos Renais/fisiologia , Morfogênese , Doenças Renais Policísticas/fisiopatologia , Transdução de Sinais , Animais , Feminino , Túbulos Renais/fisiopatologia , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA