Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 14(1): 10063, 2024 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-38698187

RESUMO

Ultra high frequency (UHF) ultrasound enables the visualization of very small structures that cannot be detected by conventional ultrasound. The utilization of UHF imaging as a new imaging technique for the 3D-in-vivo chorioallantoic membrane (CAM) model can facilitate new insights into tissue perfusion and survival. Therefore, human renal cystic tissue was grafted onto the CAM and examined using UHF ultrasound imaging. Due to the unprecedented resolution of UHF ultrasound, it was possible to visualize microvessels, their development, and the formation of anastomoses. This enabled the observation of anastomoses between human and chicken vessels only 12 h after transplantation. These observations were validated by 3D reconstructions from a light sheet microscopy image stack, indocyanine green angiography, and histological analysis. Contrary to the assumption that the nutrient supply of the human cystic tissue and the gas exchange happens through diffusion from CAM vessels, this study shows that the vasculature of the human cystic tissue is directly connected to the blood vessels of the CAM and perfusion is established within a short period. Therefore, this in-vivo model combined with UHF imaging appears to be the ideal platform for studying the effects of intravenously applied therapeutics to inhibit renal cyst growth.


Assuntos
Membrana Corioalantoide , Rim Policístico Autossômico Dominante , Ultrassonografia , Animais , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/diagnóstico por imagem , Humanos , Rim Policístico Autossômico Dominante/diagnóstico por imagem , Ultrassonografia/métodos , Galinhas , Rim/diagnóstico por imagem , Rim/irrigação sanguínea , Imageamento Tridimensional/métodos
3.
STAR Protoc ; 4(1): 101874, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36856763

RESUMO

Collecting-duct-derived renal epithelial cells switch from tubule to cyst formation; however, the cysts still form tubules after injury of the cyst-lining epithelium. Here, we provide a protocol that describes in vitro cyst growth with focus on glass-capillary-induced cyst wall injury to induce tubule formation. We detail steps for the establishment of the in vitro cyst assay, followed by puncture of the cysts in the collagen matrix. We further describe live imaging and steps to analyze the tubule growth. For complete details on the use and execution of this protocol, please refer to Scholz et al. (2022).1.


Assuntos
Cistos , Células Epiteliais , Humanos , Epitélio , Colágeno
4.
Kidney Int ; 103(5): 917-929, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36804411

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) mainly results from mutations in the PKD1 gene, which encodes polycystin 1. It is the most common inherited kidney disease and is characterized by a progressive bilateral increase in cyst number and size, often leading to kidney failure. The cellular energy sensor and regulator adenosine monophosphate stimulated protein kinase (AMPK) has been implicated as a promising new therapeutic target. To address this hypothesis, we determined the effects of a potent and selective clinical stage direct allosteric AMPK activator, PXL770, in canine and patient-derived 3D cyst models and an orthologous mouse model of ADPKD. PXL770 induced AMPK activation and dose-dependently reduced cyst growth in principal-like Madin-Darby Canine Kidney cells stimulated with forskolin and kidney epithelial cells derived from patients with ADPKD stimulated with desmopressin. In an inducible, kidney epithelium-specific Pkd1 knockout mouse model, PXL770 produced kidney AMPK pathway engagement, prevented the onset of kidney failure (reducing blood urea by 47%), decreased cystic index by 26% and lowered the kidney weight to body weight ratio by 35% compared to untreated control Pkd1 knockout mice. These effects were accompanied by a reduction of markers of cell proliferation (-48%), macrophage infiltration (-53%) and tissue fibrosis (-37%). Thus, our results show the potential of direct allosteric AMPK activation in the treatment of ADPKD and support the further development of PXL770 for this indication.


Assuntos
Cistos , Rim Policístico Autossômico Dominante , Insuficiência Renal , Camundongos , Animais , Cães , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Monofosfato de Adenosina/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Rim/metabolismo , Camundongos Knockout , Insuficiência Renal/metabolismo , Progressão da Doença , Cistos/tratamento farmacológico , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
5.
iScience ; 25(6): 104359, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35620436

RESUMO

Autosomal dominant polycystic kidney disease is the most common monogenic disease that causes end-stage renal failure. It primarily results from mutations in the PKD1 gene that encodes for Polycystin-1. How loss of Polycystin-1 translates into bilateral renal cyst development is mostly unknown. cAMP is significantly involved in cyst enlargement but its role in cyst initiation has remained elusive. Deletion of Polycystin-1 in collecting duct cells resulted in a switch from tubule to cyst formation and was accompanied by an increase in cAMP. Pharmacological elevation of cAMP in Polycystin-1-competent cells caused cyst formation, impaired plasticity, nondirectional migration, and mis-orientation, and thus strongly resembled the phenotype of Polycystin-1-deficient cells. Mis-orientation of developing tubule cells in metanephric kidneys upon loss of Polycystin-1 was phenocopied by pharmacological increase of cAMP in wildtype kidneys. In vitro, cAMP impaired tubule formation after capillary-induced injury which was further impaired by loss Polycystin-1.

6.
FASEB J ; 35(10): e21897, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34473378

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the development of bilateral renal cysts which enlarge continuously, leading to compression of adjacent intact nephrons. The growing cysts lead to a progressive decline in renal function. Cyst growth is driven by enhanced cell proliferation and chloride secretion into the cyst lumen. Chloride secretion is believed to occur mainly by the cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR), with some contribution by the calcium-activated chloride channel TMEM16A. However, our previous work suggested TMEM16A as a major factor for renal cyst formation. The contribution of CFTR to cyst formation has never been demonstrated in an adult ADPKD mouse model. We used mice with an inducible tubule-specific Pkd1 knockout, which consistently develop polycystic kidneys upon deletion of Pkd1. Cellular properties, ion currents, and cyst development in these mice were compared with that of mice carrying a co-deletion of Pkd1 and Cftr. Knockout of Cftr did not reveal any significant impact on cyst formation in the ADPKD mouse model. Furthermore, knockout of Cftr did not attenuate the largely augmented cell proliferation observed in Pkd1 knockout kidneys. Patch clamp analysis on primary renal epithelial cells lacking expression of Pkd1 indicated an only marginal contribution of CFTR to whole cell Cl- currents, which were clearly dominated by calcium-activated TMEM16A currents. In conclusion, CFTR does not essentially contribute to renal cyst formation in mice caused by deletion of Pkd1. Enhanced cell proliferation and chloride secretion is caused primarily by upregulation of the calcium-activated chloride channel TMEM16A.


Assuntos
Anoctamina-1/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Cistos/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Anoctamina-1/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cistos/genética , Cistos/patologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP/genética
7.
J Mol Med (Berl) ; 98(11): 1547-1559, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32885302

RESUMO

Progressive cyst growth leads to decline of renal function in polycystic kidney disease. Macrophage migration inhibitory factor (MIF) was found to be upregulated in cyst-lining cells in a mouse model of polycystic kidney disease and to promote cyst growth. In addition, MIF can be secreted by tubular cells and may contribute to cyst growth in an autocrine manner. However, the underlying mechanisms leading to induction of MIF in cyst-lining cells remained elusive. Here, we demonstrate that hypoxia-inducible transcription factor (HIF) 1α upregulates MIF in cyst-lining cells in a tubule-specific PKD1 knockout mouse. Pharmacological stabilization of HIF-1α resulted in significant increase of MIF in cyst epithelial cells whereas tubule-specific knockout of HIF-1α prevented MIF upregulation. Identical regulation could be found for ABCA1, which has been shown to act as a transport protein for MIF. Furthermore, we show that MIF and ABCA1 are direct target genes of HIF-1α in human primary tubular cells. Next to HIF-1α and hypoxia, we found MIF being additionally regulated by cAMP which is a strong promotor of cyst growth. In line with these findings, HIF-1α- and cAMP-dependent in vitro cyst growth could be decreased by the MIF-inhibitor ISO-1 which resulted in reduced cyst cell proliferation. In conclusion, HIF-1α and cAMP regulate MIF in primary tubular cells and cyst-lining epithelial cells, and MIF promotes cyst growth in the absence of macrophages. In line with these findings, the MIF inhibitor ISO-1 attenuates HIF-1α- and cAMP-dependent in vitro cyst enlargement. KEY MESSAGES: • MIF is upregulated in cyst-lining cells in a polycystic kidney disease mouse model. • MIF upregulation is mediated by hypoxia-inducible transcription factor (HIF) 1α. • ABCA1, transport protein for MIF, is also regulated by HIF-1α in vitro and in vivo. • MIF is additionally regulated by cAMP, a strong promotor of cyst growth. • MIF-inhibitor ISO-1 reduces HIF-1α- and cAMP-dependent cyst growth.


Assuntos
AMP Cíclico/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxirredutases Intramoleculares/metabolismo , Doenças Renais Císticas/etiologia , Doenças Renais Císticas/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Biomarcadores , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Relação Dose-Resposta a Droga , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/genética , Isoxazóis/farmacologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos , Camundongos Knockout
8.
BMC Nephrol ; 21(1): 104, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32204691

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by systemic thrombotic microangiopathy (TMA) reflected by hemolysis, anemia, thrombocytopenia and systemic organ injury. The optimal management of aHUS-patients when undergoing kidney transplantation to prevent recurrence in the allograft is eculizumab, an approved recombinant antibody targeting human complement component C5. CASE PRESENTATION: A 39 year-old woman presented with severe abdominal pain, diarrhea and emesis for 3 days. In her past medical history she had experienced an episode of aHUS leading to end stage renal disease (ESRD) in 2007 and a genetic workup revealed a heterozygous mutation in the membrane cofactor protein gene. In 2014 she underwent cadaveric kidney transplantation. Four years later she had to go back on hemodialysis due to allograft failure following a severe systemic cytomegalovirus infection resulting in transplant failure. At presentation she still received calcineurin-inhibitor therapy and reported subfebrile temperatures and pain projecting over the transplant prior to the current symptoms. A contrast enhanced CT-scan of the abdomen revealed inflammatory wall thickening of the small intestine. Diagnostic endoscopy discovered fresh blood in the small intestine without a clear source of bleeding. Histopathology of the small intestine biopsies showed severe thrombotic microangiopathy. Of note, the patient persistently had no signs of systemic hemolysis. Since the TMA of the small intestine was most likely due to aHUS, eculizumab treatment was initiated which abolished the symptoms. CONCLUSION: Here we report a patient with thrombotic microangiopathy with predominant manifestation in a single organ, the small intestine, due to aHUS with absence of systemic signs and symptoms. aHUS patients usually require a secondary trigger for the disease to manifest. In this case, the trigger may be attributed to the dysfunctional renal transplant, which was subsequently explanted. Histology of the explanted kidney showed severe inflammation due to purulent nephritis and signs of cellular rejection. After nephrectomy, we continued eculizumab therapy until the patient completely recovered. No signs of TMA recurred after discontinuation of eculizumab, further supporting the concept of the renal transplant as the main trigger of TMA of the small intestine in our patient.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Síndrome Hemolítico-Urêmica Atípica , Rejeição de Enxerto , Intestino Delgado , Falência Renal Crônica/cirurgia , Transplante de Rim , Microangiopatias Trombóticas , Adulto , Síndrome Hemolítico-Urêmica Atípica/complicações , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/fisiopatologia , Biópsia/métodos , Inativadores do Complemento/administração & dosagem , Endoscopia Gastrointestinal/métodos , Feminino , Rejeição de Enxerto/complicações , Rejeição de Enxerto/diagnóstico , Humanos , Intestino Delgado/irrigação sanguínea , Intestino Delgado/diagnóstico por imagem , Intestino Delgado/patologia , Falência Renal Crônica/etiologia , Transplante de Rim/efeitos adversos , Transplante de Rim/métodos , Proteína Cofatora de Membrana/genética , Mutação , Diálise Renal/métodos , Microangiopatias Trombóticas/diagnóstico , Microangiopatias Trombóticas/tratamento farmacológico , Microangiopatias Trombóticas/fisiopatologia , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento
9.
Cell Signal ; 69: 109524, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31904413

RESUMO

Kidney cyst growth in ADPKD is associated with regional hypoxia, presumably due to a mismatch between enlarged cysts and the peritubular capillary blood supply and compression of peritubular capillaries in cyst walls. Regional hypoxia leads to activation of hypoxia-inducible transcription factors, with the two main HIF isoforms, HIF-1 and HIF-2 expressed in cyst epithelia and pericystic interstitial cells, respectively. While HIF-2 activation is linked to EPO production, mitigating the anemia that normally accompanies chronic kidney disease, HIF-1 promotes cyst growth. HIF-dependent cyst growth is primarily due to an increase in chloride-dependent fluid secretion into the cyst lumen. However, given the broad spectrum of HIF-target genes, additional HIF-mediated pathways may also contribute to cyst progression. Furthermore, hypoxia can influence cyst growth through the generation of reactive oxygen species. Since cyst expansion aggravates regional hypoxia, a feedforward loop is established that accelerates cyst expansion and disease progression. Inhibiting the HIF pathway and/or HIF target genes that are of particular relevance for HIF-dependent cyst fluid secretion may therefore represent novel therapeutic approaches to retard the progression of APDKD.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Oxigênio/metabolismo , Rim Policístico Autossômico Dominante , Animais , Cistos/metabolismo , Cistos/patologia , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Rim/metabolismo , Rim/patologia , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia
10.
Kidney Int ; 94(5): 887-899, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30173898

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by mutations of the PKD1 gene and characterized by growth of bilateral renal cysts. Cyst growth is accompanied by regional hypoxia and induction of hypoxia-inducible factor (HIF)-1α in cyst-lining epithelial cells. To determine the relevance of HIF-1α for cyst growth in vivo we used an inducible kidney epithelium-specific knockout mouse to delete Pkd1 at postnatal day 20 or 35 to induce polycystic kidney disease of different severity and analyzed the effects of Hif-1α co-deletion and HIF-1α stabilization using a prolyl-hydroxylase inhibitor. HIF-1α expression was enhanced in kidneys with progressive cyst growth induced by early Pkd1 deletion, but unchanged in the milder phenotype induced by later Pkd1 deletion. Hif-1α co-deletion significantly attenuated cyst growth in the severe, but not in the mild, phenotype. Application of a prolyl-hydroxylase inhibitor resulted in severe aggravation of the mild phenotype with rapid loss of renal function. HIF-1α expression was associated with induction of genes that mediate calcium-activated chloride secretion. Thus, HIF-1α does not seem to play a role in early cyst formation, but accelerates cyst growth during progressive polycystic kidney disease. This novel mechanism of cyst growth may qualify as a therapeutic target.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Rim Policístico Autossômico Dominante/etiologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Rim Policístico Autossômico Dominante/terapia
11.
Am J Physiol Renal Physiol ; 315(6): F1777-F1786, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30156115

RESUMO

TMEM16A is a transmembrane protein from a conserved family of calcium-activated proteins that is highly expressed in the kidney. TMEM16A confers calcium-activated chloride channel activity, which is of importance for various cellular functions in secretory epithelia and involved in secretion-dependent renal cyst growth. However, its specific function in renal physiology has remained elusive so far. Therefore, we generated conditional nephron-specific TMEM16A-knockout mice and found that these animals suffered from albuminuria. Kidney histology demonstrated an intact corticomedullary differentiation and absence of cysts. Electron microscopy showed a normal slit diaphragm. However, the total number of glomeruli and total nephron count was decreased in TMEM16A-knockout animals. At the same time, glomerular diameter was increased, presumably as a result of the hyperfiltration in the remaining glomeruli. TUNEL and PCNA stainings showed increased cell death and increased proliferation. Proximal tubular cilia were intact in young animals, but the number of properly ciliated cells was decreased in older, albuminuric animals. Taken together, our data suggest that TMEM16A may be involved in ureteric bud branching and proper nephron endowment. Loss of TMEM16A resulted in reduced nephron number and, subsequently, albuminuria and tubular damage.


Assuntos
Albuminúria/metabolismo , Anoctamina-1/deficiência , Glomerulonefrite/metabolismo , Glomérulos Renais/metabolismo , Túbulos Renais Proximais/metabolismo , Albuminúria/genética , Albuminúria/patologia , Albuminúria/fisiopatologia , Animais , Anoctamina-1/genética , Anoctamina-1/metabolismo , Morte Celular , Proliferação de Células , Cílios/metabolismo , Cílios/ultraestrutura , Predisposição Genética para Doença , Taxa de Filtração Glomerular , Glomerulonefrite/genética , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Humanos , Glomérulos Renais/fisiopatologia , Glomérulos Renais/ultraestrutura , Túbulos Renais Proximais/fisiopatologia , Túbulos Renais Proximais/ultraestrutura , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenótipo
12.
Purinergic Signal ; 12(4): 687-695, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27565965

RESUMO

Polycystic kidney diseases are characterized by numerous renal cysts that continuously enlarge resulting in compression of intact nephrons and tissue hypoxia. Recently, we have shown that hypoxia-inducible factor (HIF)-1α promotes secretion-dependent cyst expansion, presumably by transcriptional regulation of proteins that are involved in calcium-activated chloride secretion. Here, we report that HIF-1α directly activates expression of the purinergic receptor P2Y2R in human primary renal tubular cells. In addition, we found that P2Y2R is highly expressed in cyst-lining cells of human ADPKD kidneys as well as PKD1 orthologous mouse kidneys. Knockdown of P2Y2R in renal collecting duct cells inhibited calcium-dependent chloride secretion in Ussing chamber analyses. In line with these findings, knockdown of P2Y2R retarded cyst expansion in vitro and prevented ATP- and HIF-1α-dependent cyst growth. In conclusion, P2Y2R mediates ATP-dependent cyst growth and is transcriptionally regulated by HIF-1α. These findings provide further mechanistic evidence on how hypoxia promotes cyst growth.


Assuntos
Cistos/metabolismo , Células Epiteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Túbulos Renais Proximais/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Animais , Cistos/patologia , Células Epiteliais/citologia , Feminino , Humanos , Túbulos Renais Proximais/citologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade
13.
Curr Opin Nephrol Hypertens ; 25(3): 180-6, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27023836

RESUMO

PURPOSE OF REVIEW: Kidney development depends on outgrowth of the ureteric bud into the metanephric mesenchyme. The number of ureteric bud branching events determines the final number of nephrons, which correlates inversely with the risk for development of chronic kidney disease and arterial hypertension during lifetime. The purpose of this review is to highlight the influence of oxygen on nephrogenesis and to describe cellular mechanisms by which hypoxia can impair nephron formation. RECENT FINDINGS: Although kidney development normally takes place under hypoxic conditions, nephrogenesis is impaired when oxygen availability falls below the usual range. Hypoxia-inducible factors (HIF) play an important role in linking low oxygen concentrations to the biology of nephron formation, but their effect appears to be cell type dependent. In ureteric bud cells, HIF stimulates tubulogenesis, whereas HIF stabilization in cells of the metanephric mesenchyme results in secretion of growth factors, including vascular endothelial growth factor A, which in aggregate inhibit ureteric bud branching. The balance between pro and antibranching effects may be altered in various ways, but the inhibitory effect usually seems to predominate under reduced oxygen concentrations, explaining how intrauterine hypoxia can lead to low nephron numbers. SUMMARY: Oxygen availability has a complex influence on nephrogenesis. Oxygen concentrations outside an optimal low range may affect nephron endowment. Associations between placental insufficiency and increased risk for chronic kidney disease and arterial hypertension during later life may to a large extent be due to direct effects of reduced oxygen supply to the metanephric mesenchyme and mediated through the HIF pathway.


Assuntos
Hipóxia/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Morfogênese/fisiologia , Néfrons/metabolismo , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
J Mol Med (Berl) ; 94(1): 107-17, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26334260

RESUMO

UNLABELLED: Polycystic kidney diseases are characterized by the development of numerous bilateral renal cysts that continuously enlarge resulting in a decline of kidney function due to compression of intact nephrons. Cyst growth is driven by transepithelial chloride secretion which depends on both intracellular cAMP and calcium. Mechanisms that are involved in the regulation of the underlying secretory pathways remain incompletely understood. Here we show that glucose concentration has a strong impact on cyst growth of renal tubular cells within a collagen matrix as well as in embryonic kidneys deficient or competent for Pkd1. Glucose-dependent cyst growth correlates with the transcriptional induction of the calcium-activated chloride channel anoctamin 1 (ANO1) and its increased expression in the apical membrane of cyst-forming cells. Inhibition of ANO1 with the specific inhibitor CaCCinh-AO1 significantly decreases glucose-dependent cyst growth in both models. Ussing chamber analyses revealed increased apical chloride secretion of renal tubular cells upon exposure to high glucose medium which can also be inhibited by the use of CaCCinh-AO1. These data suggest that glycemic control may help to reduce renal cyst growth in patients with polycystic kidney disease. KEY MESSAGE: Renal cyst growth depends on glucose concentration in two in vitro cyst models. High glucose leads to upregulation of the calcium-activated chloride channel ANO1. High glucose promotes calcium-activated chloride secretion via ANO1. Glucose-dependent secretion can be inhibited by a specific inhibitor of ANO1.


Assuntos
Cloretos/metabolismo , Cistos/patologia , Glucose/farmacologia , Túbulos Renais/patologia , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP/genética , Animais , Anoctamina-1 , Cálcio/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/biossíntese , Canais de Cloreto/genética , AMP Cíclico/metabolismo , Cães , Túbulos Renais/citologia , Túbulos Renais/embriologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação Transcricional/genética
15.
J Am Soc Nephrol ; 25(3): 465-74, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24203996

RESUMO

Polycystic kidney diseases are characterized by numerous bilateral renal cysts that continuously enlarge and, through compression of intact nephrons, lead to a decline in kidney function over time. We previously showed that cyst enlargement is accompanied by regional hypoxia, which results in the stabilization of hypoxia-inducible transcription factor-1α (HIF-1α) in the cyst epithelium. Here we demonstrate a correlation between cyst size and the expression of the HIF-1α-target gene, glucose transporter 1, and report that HIF-1α promotes renal cyst growth in two in vitro cyst models-principal-like MDCK cells (plMDCKs) within a collagen matrix and cultured embryonic mouse kidneys stimulated with forskolin. In both models, augmenting HIF-1α levels with the prolyl hydroxylase inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate enhanced cyst growth. In addition, inhibition of HIF-1α degradation through tubule-specific knockdown of the von Hippel-Lindau tumor suppressor increased cyst size in the embryonic kidney cyst model. In contrast, inhibition of HIF-1α by chetomin and knockdown of HIF-1α both decreased cyst growth in these models. Consistent with previous reports, plMDCK cyst enlargement was driven largely by transepithelial chloride secretion, which consists, in part, of a calcium-activated chloride conductance. plMDCKs deficient for HIF-1α almost completely lacked calcium-activated chloride secretion. We conclude that regional hypoxia in renal cysts contributes to cyst growth, primarily due to HIF-1α-dependent calcium-activated chloride secretion. These findings identify the HIF system as a novel target for inhibition of cyst growth.


Assuntos
Cloretos/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Doenças Renais Policísticas/etiologia , Animais , Canais de Cloreto/metabolismo , Cães , Feminino , Regulação da Expressão Gênica , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Hipóxia/fisiopatologia , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos C57BL , Doenças Renais Policísticas/metabolismo
16.
Kidney Int ; 85(5): 1058-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24152967

RESUMO

Polycystic kidney diseases are characterized by multiple bilateral renal cysts that gradually enlarge and lead to a decline in renal function. Cyst enlargement is driven by transepithelial chloride secretion, stimulated by enhanced levels of cyclic adenosine monophosphate, which activates apical cystic fibrosis transmembrane conductance regulator chloride channels. However, chloride secretion by calcium-dependent chloride channels, activated through stimulation of purinergic receptors, also has a major impact. To identify the molecular basis of calcium-dependent chloride secretion in cyst expansion, we determined the role of anoctamin 1 and 6, two recently discovered calcium-activated chloride channels both of which are expressed in epithelial cells. We found that anoctamin 1, which plays a role in epithelial fluid secretion and proliferation, is strongly expressed in principal-like MDCK cells (PLCs) forming cysts within a collagen matrix, in an embryonic kidney cyst model, and in human autosomal dominant polycystic kidney disease tissue. Knockdown of anoctamin 1 but not anoctamin 6 strongly diminished the calcium-dependent chloride secretion of PLCs. Moreover, two inhibitors of anoctamin ion channels, tannic acid and a more selective inhibitor of anoctamin 1, significantly inhibited PLC cyst growth and cyst enlargement in an embryonic kidney cyst model. Knockdown of ANO1 by morpholino analogs also attenuated embryonic cyst growth. Thus, calcium-activated chloride secretion by anoctamin 1 appears to be a crucial component of renal cyst growth.


Assuntos
Proliferação de Células , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Proteínas de Neoplasias/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Animais , Anoctamina-1 , Proliferação de Células/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Progressão da Doença , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Humanos , Rim/efeitos dos fármacos , Rim/embriologia , Rim/patologia , Células Madin Darby de Rim Canino , Masculino , Moduladores de Transporte de Membrana/farmacologia , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Rim Policístico Autossômico Dominante/patologia , Agonistas Purinérgicos/farmacologia , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção
17.
Nephrol Dial Transplant ; 26(11): 3458-65, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21804086

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure. Aberrant epithelial cell proliferation is a major cause of progressive cyst enlargement in ADPKD. Since activation of the Ras/Raf signaling system has been detected in cyst-lining epithelia, inhibition of Raf kinase has been proposed as an approach to retard the progression of ADPKD. Methods and results. PLX5568, a novel selective small molecule inhibitor of Raf kinases, attenuated proliferation of human ADPKD cyst epithelial cells. It reduced in vitro cyst growth of Madin-Darby Canine Kidney cells and of human ADPKD cells within a collagen gel. In male cy/+ rats with polycystic kidneys, PLX5568 inhibited renal cyst growth along with a significant reduction in the number of proliferating cell nuclear antigen- and phosphorylated extracellular signal-regulated kinase-positive cyst-lining epithelial cells. Furthermore, treated animals showed increased capacity to concentrate urine. However, PLX5568 did not lead to a consistent improvement of renal function. Moreover, although relative cyst volume was decreased, total kidney-to-body weight ratio was not significantly reduced by PLX5568. Further analyses revealed a 2-fold increase of renal and hepatic fibrosis in animals treated with PLX5568. CONCLUSIONS: PLX5568 attenuated cyst enlargement in vitro and in a rat model of ADPKD without improving kidney function, presumably due to increased renal fibrosis. These data suggest that effective therapies for the treatment of ADPKD will need to target fibrosis as well as the growth of cysts.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cistos/patologia , Rim/fisiopatologia , Cirrose Hepática/induzido quimicamente , Doenças Renais Policísticas/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Animais , Células Cultivadas , Cistos/tratamento farmacológico , Cães , Células Epiteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Rim/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno , Fosforilação/efeitos dos fármacos , Doenças Renais Policísticas/enzimologia , Doenças Renais Policísticas/patologia , Inibidores de Proteínas Quinases/toxicidade , Ratos , Ratos Sprague-Dawley
18.
J Mol Med (Berl) ; 89(3): 251-61, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21206992

RESUMO

It has been suggested that more than 70% of the renal cysts in patients with autosomal dominant polycystic kidney disease (ADPKD) arise from the collecting duct and that within this segment cysts originate almost exclusively from principal rather than intercalated cells. The mechanisms for this predisposition of principal cells have so far remained elusive. We, therefore, used Madin-Darby canine kidney (MDCK) subclones resembling principal cells and alpha-intercalated cells in a three-dimensional in vitro model to determine differences in cystogenesis and cyst growth, including the response to cyclic adenosine monophosphate (cAMP) elevation and the dependence on ATP signaling. We found that in vitro cysts developed only from principal-like but not from intercalated-like MDCK cell clones. This specificity could be verified in mixed MDCK cultures enriched for principal- or intercalated-like cells. In vitro cyst growth upon elevation of intracellular cAMP was mainly driven by fluid secretion, rather than increased cell proliferation. The cAMP-dependent fluid secretion was found to depend on extracellular adenosine-5'-triphosphate (ATP) and to act synergistically with purinergic signaling, as the use of the ATP scavenger apyrase, as well as the P2 receptor inhibitor suramin, reduced cAMP-driven fluid secretion, while increasing extracellular ATP potentiated cAMP-mediated cyst growth. In conclusion, we provide in vitro evidence for the ability of principal rather than intercalated cells to form cysts, based on a synergism of cAMP and ATP signaling in enhancing apical fluid secretion.


Assuntos
Trifosfato de Adenosina/metabolismo , AMP Cíclico/metabolismo , Cistos/metabolismo , Animais , Western Blotting , Linhagem Celular , Cães , Imuno-Histoquímica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA