Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Am J Surg Pathol ; 46(2): 226-232, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34889853

RESUMO

Superficial angiomyxomas (SAMs) are benign cutaneous tumors that arise de novo and in the setting of the Carney complex (CC), an autosomal dominant disease with several cutaneous manifestations including lentigines and pigmented epithelioid melanocytomas. Although most SAM do not pose a diagnostic challenge, a subset can demonstrate histopathologic overlap with other myxoid tumors that arise in the skin and subcutis. Traditional immunohistochemical markers are of limited utility when discriminating SAM from histopathologic mimics. Since protein kinase A regulatory subunit 1 alpha (PRKAR1A) genetic alterations underlie most CC cases, we investigated whether SAM demonstrate loss of PRKAR1A protein expression by immunohistochemistry. In our series, 29 SAM, 26 myxofibrosarcoma, 5 myxoid dermatofibrosarcoma protuberans, 11 superficial acral fibromyxomas, and 18 digital mucous cysts were characterized. Of the 29 SAM examined in this study, 1 was associated with documented CC in a 5-year-old girl. SAM tended to arise in adults (mean 49.7 y; range: 5 to 87 y). Loss of PRKAR1A was seen in 55.2% of cases (16/29) and had a male predilection (87.5%, 12/16). PRKAR1A-inactivated SAM demonstrated significant nuclear enlargement (100%, 16/16 vs. 23.1%, 3/13), multinucleation (81.3%, 13/16 vs. 23.1%, 3/13), and presence of neutrophils (43.8%, 7/16 vs. 0%, 0/13). In contrast, PRKAR1A was retained in all cases of myxofibrosarcoma (100%, 26/26), myxoid dermatofibrosarcoma protuberans (100%, 5/5), superficial acral fibromyxomas (100%, 11/11), and digital mucous cyst (100%, 18/18). Taken together, PRKAR1A loss by immunohistochemistry can be used as an adjunctive assay to support the diagnosis of SAM given the high specificity of this staining pattern compared with histopathologic mimics.


Assuntos
Biomarcadores Tumorais/deficiência , Complexo de Carney/enzimologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Cistos/enzimologia , Dermatofibrossarcoma/enzimologia , Fibroma/enzimologia , Imuno-Histoquímica , Mixoma/enzimologia , Neoplasias Cutâneas/enzimologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Complexo de Carney/patologia , Criança , Pré-Escolar , Cistos/patologia , Dermatofibrossarcoma/patologia , Feminino , Fibroma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mixoma/patologia , Valor Preditivo dos Testes , Neoplasias Cutâneas/patologia
2.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-34638853

RESUMO

DNA-dependent protein kinase (DNA-PK) is a serine/threonine protein involved in DNA damage response (DDR) signaling that may mediate kidney cyst growth in autosomal dominant polycystic kidney disease (ADPKD) due to its pleiotropic effects on proliferation and survival. To test this hypothesis, the expression of DNA-PK in human ADPKD and the in vitro effects of DNA-PK inhibition in a three-dimensional model of Madin-Darby Canine Kidney (MDCK) cyst growth and human ADPKD cells were assessed. In human ADPKD, the mRNA expression for all three subunits of the DNA-PK complex was increased, and using immunohistochemistry, the catalytic subunit (DNA-PKcs) was detected in the cyst lining epithelia of human ADPKD, in a focal manner. In vitro, NU7441 (a DNA-PK kinase inhibitor) reduced MDCK cyst growth by up to 52% after long-term treatment over 6-12 days. Although human ADPKD cell lines (WT9-7/WT9-12) did not exhibit synthetic lethality in response to DNA-PK kinase inhibition compared to normal human kidney cells (HK-2), the combination of low-dose NU7441 enhanced the anti-proliferative effects of sirolimus in WT9-7 and WT9-12 cells by 17 ± 10% and 11 ± 7%, respectively. In conclusion, these preliminary data suggest that DNA-PK mediates kidney cyst growth in vivo without a synthetically lethal interaction, conferring cell-specificity in human ADPKD cells. NU7441 enhanced the anti-proliferative effects of rapamycin complex 1 inhibitors, but the effect was modest.


Assuntos
Cistos/genética , Proteína Quinase Ativada por DNA/genética , Perfilação da Expressão Gênica/métodos , Rim/metabolismo , Rim Policístico Autossômico Dominante/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cromonas/farmacologia , Cistos/tratamento farmacológico , Cistos/enzimologia , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteína Quinase Ativada por DNA/metabolismo , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Rim/patologia , Células Madin Darby de Rim Canino , Morfolinas/farmacologia , Rim Policístico Autossômico Dominante/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/genética
3.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G464-G473, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33439105

RESUMO

Polycystic liver disease (PLD) is a hereditary liver disease in which the number of cysts increases over time, causing various abdominal symptoms and poor quality of life. Although effective treatment for PLD has not been established, we recently reported that long-term exercise ameliorated liver cyst formation and fibrosis with the activation of AMP-activated protein kinase (AMPK) in polycystic kidney (PCK) rats, a PLD model. Therefore, the aim of this study was to investigate whether metformin, an indirect AMPK activator, was effective in PCK rats. PCK rats were randomly divided into a control (Con) group and a metformin-treated (Met) group. The Met group was treated orally with metformin in drinking water. After 12 wk, liver function, histology, and signaling cascades of PLD were examined in the groups. Metformin did not affect the body weight or liver weight, but it reduced liver cyst formation, cholangiocyte proliferation, and fibrosis around the cyst. Metformin increased the phosphorylation of AMPK and tuberous sclerosis complex 2 and decreased the phosphorylation of mammalian target of rapamycin, S6, and extracellular signal-regulated kinase and the expression of cystic fibrosis transmembrane conductance regulator, aquaporin I, transforming growth factor-ß, and type 1 collagen without changes in apoptosis or collagen degradation factors in the liver. Metformin slows the development of cyst formation and fibrosis with the activation of AMPK and inhibition of signaling cascades responsible for cellular proliferation and fibrosis in the liver of PCK rats.NEW & NOTEWORTHY This study indicates that metformin, an indirect AMPK activator slows liver cyst formation and fibrosis in PLD rat model. Metformin attenuates excessive cell proliferation in the liver with the inactivation of mTOR and ERK pathways. Metformin also reduces the expression of proteins responsible for cystic fluid secretion and liver fibrosis. Metformin and AMPK activators may be potent drugs for polycystic liver disease.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proliferação de Células/efeitos dos fármacos , Cistos/tratamento farmacológico , Ativadores de Enzimas/farmacologia , Cirrose Hepática Experimental/tratamento farmacológico , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Metformina/farmacologia , Animais , Cistos/enzimologia , Cistos/patologia , Progressão da Doença , Ativação Enzimática , Fígado/enzimologia , Fígado/patologia , Cirrose Hepática Experimental/enzimologia , Cirrose Hepática Experimental/patologia , Hepatopatias/enzimologia , Hepatopatias/patologia , Masculino , Fosforilação , Ratos , Transdução de Sinais , Fatores de Tempo
4.
J Biol Chem ; 292(43): 17897-17908, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28887310

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is associated with progressive enlargement of multiple renal cysts, often leading to renal failure that cannot be prevented by a current treatment. Two proteins encoded by two genes are associated with ADPKD: PC1 (pkd1), primarily a signaling molecule, and PC2 (pkd2), a Ca2+ channel. Dysregulation of cAMP signaling is central to ADPKD, but the molecular mechanism is unresolved. Here, we studied the role of histone deacetylase 6 (HDAC6) in regulating cyst growth to test the possibility that inhibiting HDAC6 might help manage ADPKD. Chemical inhibition of HDAC6 reduced cyst growth in PC1-knock-out mice. In proximal tubule-derived, PC1-knock-out cells, adenylyl cyclase 6 and 3 (AC6 and -3) are both expressed. AC6 protein expression was higher in cells lacking PC1, compared with control cells containing PC1. Intracellular Ca2+ was higher in PC1-knock-out cells than in control cells. HDAC inhibition caused a drop in intracellular Ca2+ and increased ATP-simulated Ca2+ release. HDAC6 inhibition reduced the release of Ca2+ from the endoplasmic reticulum induced by thapsigargin, an inhibitor of endoplasmic reticulum Ca2+-ATPase. HDAC6 inhibition and treatment of cells with the intracellular Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) reduced cAMP levels in PC1-knock-out cells. Finally, the calmodulin inhibitors W-7 and W-13 reduced cAMP levels, and W-7 reduced cyst growth, suggesting that AC3 is involved in cyst growth regulated by HDAC6. We conclude that HDAC6 inhibition reduces cell growth primarily by reducing intracellular cAMP and Ca2+ levels. Our results provide potential therapeutic targets that may be useful as treatments for ADPKD.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , AMP Cíclico/metabolismo , Cistos/enzimologia , Inibidores de Histona Desacetilases/farmacocinética , Histona Desacetilases/metabolismo , Rim Policístico Autossômico Dominante/enzimologia , Animais , Quelantes de Cálcio/farmacologia , Sinalização do Cálcio/genética , Linhagem Celular , AMP Cíclico/genética , Cistos/genética , Cistos/patologia , Modelos Animais de Doenças , Desacetilase 6 de Histona , Histona Desacetilases/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Tapsigargina/farmacologia
5.
J Clin Invest ; 127(7): 2751-2764, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28604386

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is driven by mutations in PKD1 and PKD2 genes. Recent work suggests that epigenetic modulation of gene expression and protein function may play a role in ADPKD pathogenesis. In this study, we identified SMYD2, a SET and MYND domain protein with lysine methyltransferase activity, as a regulator of renal cyst growth. SMYD2 was upregulated in renal epithelial cells and tissues from Pkd1-knockout mice as well as in ADPKD patients. SMYD2 deficiency delayed renal cyst growth in postnatal kidneys from Pkd1 mutant mice. Pkd1 and Smyd2 double-knockout mice lived longer than Pkd1-knockout mice. Targeting SMYD2 with its specific inhibitor, AZ505, delayed cyst growth in both early- and later-stage Pkd1 conditional knockout mouse models. SMYD2 carried out its function via methylation and activation of STAT3 and the p65 subunit of NF-κB, leading to increased cystic renal epithelial cell proliferation and survival. We further identified two positive feedback loops that integrate epigenetic regulation and renal inflammation in cyst development: SMYD2/IL-6/STAT3/SMYD2 and SMYD2/TNF-α/NF-κB/SMYD2. These pathways provide mechanisms by which SMYD2 might be induced by cyst fluid IL-6 and TNF-α in ADPKD kidneys. The SMYD2 transcriptional target gene Ptpn13 also linked SMYD2 to other PKD-associated signaling pathways, including ERK, mTOR, and Akt signaling, via PTPN13-mediated phosphorylation.


Assuntos
Proliferação de Células , Cistos/enzimologia , Epigênese Genética , Células Epiteliais/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Benzoxazinas/farmacologia , Cistos/tratamento farmacológico , Cistos/genética , Cistos/patologia , Células Epiteliais/patologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Metilação/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Rim Policístico Autossômico Dominante , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 13/genética , Proteína Tirosina Fosfatase não Receptora Tipo 13/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
6.
Am J Physiol Renal Physiol ; 313(3): F677-F686, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28615245

RESUMO

The failure of the polycystins (PCs) to function in primary cilia is thought to be responsible for autosomal dominant polycystic kidney disease (ADPKD). Primary cilia integrate multiple cellular signaling pathways, including calcium, cAMP, Wnt, and Hedgehog, which control cell proliferation and differentiation. It has been proposed that mutated PCs result in reduced intracellular calcium, which in turn upregulates cAMP, protein kinase A (PKA) signaling, and subsequently other proliferative signaling pathways. However, the role of PKA in ADPKD has not been directly ascertained in vivo, although the expression of the main regulatory subunit of PKA in cilia and other compartments (PKA-RIα, encoded by PRKAR1A) is increased in a mouse model orthologous to ADPKD. Therefore, we generated a kidney-specific knockout of Prkar1a to examine the consequences of constitutive upregulation of PKA on wild-type and Pkd1 hypomorphic (Pkd1RC) backgrounds. Kidney-specific loss of Prkar1a induced renal cystic disease and markedly aggravated cystogenesis in the Pkd1RC models. In both settings, it was accompanied by upregulation of Src, Ras, MAPK/ERK, mTOR, CREB, STAT3, Pax2 and Wnt signaling. On the other hand, Gli3 repressor activity was enhanced, possibly contributing to hydronephrosis and impaired glomerulogenesis in some animals. To assess the relevance of these observations in humans we looked for and found evidence for kidney and liver cystic phenotypes in the Carney complex, a tumoral syndrome caused by mutations in PRKAR1A These observations expand our understanding of the pathogenesis of ADPKD and demonstrate the importance of PRKAR1A highlighting PKA as a therapeutic target in ADPKD.


Assuntos
Complexo de Carney/enzimologia , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Cistos/enzimologia , Rim/enzimologia , Hepatopatias/enzimologia , Rim Policístico Autossômico Dominante/enzimologia , Adolescente , Adulto , Animais , Complexo de Carney/diagnóstico por imagem , Complexo de Carney/genética , Proliferação de Células , Criança , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/deficiência , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Cistos/diagnóstico por imagem , Cistos/genética , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Rim/patologia , Rim/fisiopatologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Hepatopatias/diagnóstico por imagem , Hepatopatias/genética , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição PAX2/metabolismo , Fenótipo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Rim Policístico Autossômico Dominante/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Canais de Cátion TRPP/genética , Via de Sinalização Wnt , Adulto Jovem , Proteína Gli3 com Dedos de Zinco , Proteínas ras/metabolismo , Quinases da Família src/metabolismo
7.
Gut ; 63(10): 1658-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24436140

RESUMO

OBJECTIVE: Polycystic liver diseases (PCLDs) are genetic disorders characterised by progressive bile duct dilatation and/or cyst development. Their pathogenesis is a consequence of hyperproliferation, hypersecretion and microRNA alterations in cholangiocytes. Here we evaluate the role of matrix metalloproteases (MMPs) in the hepatic cystogenesis of PCLDs. DESIGN: Metalloprotease activity was measured by microfluorimetric assays in normal and polycystic cholangiocyte cultures from humans and rats, and gene expression by real time quantitative PCR. The role of cytokines, oestrogens and growth factors present in the cystic fluid of PCLD patients was evaluated for MMP activity. The MMP inhibitor marimastat was examined for cystic expansion in vitro and in polycystic kidney (PCK) rats. RESULTS: Polycystic human and rat cholangiocytes displayed increased MMP activity, which was associated with increased mRNA levels of different MMPs. Interleukin (IL)-6 and IL-8, and 17ß-oestradiol, all stimulated MMP activity in human cholangiocytes. The presence of antibodies against IL-6 and/or IL-8 receptor/s inhibited baseline MMP hyperactivity of polycystic human cholangiocytes but had no effect on normal human cholangiocytes. MMP-3 was overexpressed in cystic cholangiocytes from PCLD human and PCK rat livers by immunohistochemistry. Marimastat reduced MMP hyperactivity of polycystic human and rat cholangiocytes and blocked the cystic expansion of PCK cholangiocytes cultured in three-dimensions. Chronic treatment of 8-week-old PCK rats with marimastat inhibited hepatic cystogenesis and fibrosis. CONCLUSIONS: PCLDs are associated with cholangiocyte MMP hyperactivity resulting from autocrine/paracrine stimulation by IL-6 and IL-8. Inhibition of this MMP hyperactivity with marimastat decreased hepatic cystogenesis in vitro and in an animal model of PCLD, offering a potential therapeutic tool.


Assuntos
Ductos Biliares/enzimologia , Cistos/prevenção & controle , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Hepatopatias/prevenção & controle , Metaloendopeptidases/antagonistas & inibidores , Animais , Ductos Biliares/patologia , Western Blotting , Técnicas de Cultura de Células , Cistos/enzimologia , Citocinas/metabolismo , Citofotometria , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Imuno-Histoquímica , Fígado/patologia , Hepatopatias/enzimologia , Masculino , Ratos , Reação em Cadeia da Polimerase em Tempo Real
8.
J Laryngol Otol ; 127(5): 479-82, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23575386

RESUMO

OBJECTIVE: We investigated lactate dehydrogenase isoenzyme patterns in the cyst fluid of auricular pseudocysts and autogenous blood, to assist the diagnosis of auricular pseudocyst. METHODS: Twenty patients with auricular pseudocysts participated in this study conducted in Kaohsiung Medical University Hospital between February 2007 and June 2010. Patterns of lactate dehydrogenase in cyst fluid and autogenous blood were analysed. RESULTS: Levels of lactate dehydrogenase 1 and 2 were lower in auricular pseudocysts than in autogenous blood, whereas levels of lactate dehydrogenase 4 and 5 were higher; this difference was statistically significant (p < 0.001). CONCLUSION: Lactate dehydrogenase isoenzyme patterns in auricular pseudocyst fluid indicated higher percentage distributions of lactate dehydrogenase 4 and 5 and lower percentage distributions of lactate dehydrogenase 1 and 2. An effective laboratory method of evaluating the different lactate dehydrogenase isoenzyme components was developed; this method may improve the accuracy of auricular pseudocyst diagnosis.


Assuntos
Líquido Cístico/química , Líquido Cístico/enzimologia , Cistos/química , Cistos/enzimologia , Pavilhão Auricular , Otopatias/enzimologia , L-Lactato Desidrogenase/análise , Adulto , Feminino , Humanos , Isoenzimas/análise , Masculino , Estudos Prospectivos
9.
Hepatology ; 56(6): 2363-74, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22653837

RESUMO

UNLABELLED: Mutations in polycystins are a cause of polycystic liver disease. In polycystin-2 (PC2)-defective mice, cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent activation of the Rat Sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen signal-regulated kinase-extracellular signal-regulated kinase (ERK) 1/2 pathway stimulates the growth of liver cysts. To test the hypothesis that sorafenib, a Raf inhibitor used for the treatment of liver and kidney cancers, inhibits liver cyst growth in PC2-defective mice, we treated PC2 (i.e., Pkd2(flox/-) :pCxCreER(TM) [Pkd2cKO]) mice with sorafenib-tosylate for 8 weeks (20-60 mg/kg/day). Sorafenib caused an unexpected increase in liver cyst area, cell proliferation (Ki67), and expression of phosphorylated ERK (pERK) compared with Pkd2cKO mice treated with vehicle. When given to epithelial cells isolated from liver cysts of Pkd2cKO mice (Pkd2cKO-cells), sorafenib progressively stimulated pERK1/2 and cell proliferation [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and bromodeoxyuridine assay (MTS)] at doses between 0.001 and 1 µM; however, both pERK1/2 and cell proliferation significantly decreased at the dose of 10 µM. Raf kinase activity assay showed that whereas B-Raf is inhibited by sorafenib in both wild-type (WT) and Pkd2cKO cells, Raf-1 is inhibited in WT cells but is significantly stimulated in Pkd2cKO cells. In Pkd2cKO cells pretreated with the PKA inhibitor 14-22 amide, myristolated (1 µM) and in mice treated with octreotide in combination with sorafenib, the paradoxical activation of Raf/ERK1/2 was abolished, and cyst growth was inhibited. CONCLUSION: In PC2-defective cells, sorafenib inhibits B-Raf but paradoxically activates Raf-1, resulting in increased ERK1/2 phosphorylation, cell proliferation, and cyst growth in vivo. These effects are consistent with the ability of Raf inhibitors to transactivate Raf-1 when a PKA-activated Ras promotes Raf-1/B-Raf heterodimerization, and are inhibited by interfering with cAMP/PKA signaling both in vitro and in vivo, as shown by the reduction of liver cysts in mice treated with combined octreotide and sorafenib.


Assuntos
Benzenossulfonatos/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Cistos/enzimologia , Hepatopatias/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Canais de Cátion TRPP/deficiência , Animais , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Benzenossulfonatos/uso terapêutico , Ductos Biliares/citologia , Ductos Biliares/metabolismo , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cistos/tratamento farmacológico , Cistos/patologia , Quimioterapia Combinada , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Antígeno Ki-67/metabolismo , Hepatopatias/tratamento farmacológico , Hepatopatias/patologia , Camundongos , Camundongos Knockout , Niacinamida/análogos & derivados , Octreotida/farmacologia , Octreotida/uso terapêutico , Compostos de Fenilureia , Fosforilação , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Piridinas/uso terapêutico , Sorafenibe , Canais de Cátion TRPP/genética
10.
Gastroenterology ; 142(3): 622-633.e4, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155366

RESUMO

BACKGROUND & AIMS: In polycystic kidney disease and polycystic liver disease (PLD), the normally nonproliferative hepato-renal epithelia acquire a proliferative, cystic phenotype that is linked to overexpression of cell division cycle 25 (Cdc25)A phosphatase and cell-cycle deregulation. We investigated the effects of Cdc25A inhibition in mice and rats via genetic and pharmacologic approaches. METHODS: Cdc25A(+/-) mice (which have reduced levels of Cdc25A) were cross-bred with polycystic kidney and hepatic disease 1 (Pkhd1(del2/del2)) mice (which have increased levels of Cdc25A and develop hepatic cysts). Cdc25A expression was analyzed in livers of control and polycystic kidney (PCK) rats, control and polycystic kidney 2 (Pkd2(ws25/-)) mice, healthy individuals, and patients with PLD. We examined effects of pharmacologic inhibition of Cdc25A with vitamin K3 (VK3) on the cell cycle, proliferation, and cyst expansion in vitro; hepato-renal cystogenesis in PCK rats and Pkd2(ws25/-)mice; and expression of Cdc25A and the cell-cycle proteins regulated by Cdc25A. We also examined the effects of the Cdc25A inhibitor PM-20 on hepato-renal cystogenesis in Pkd2(ws25/-) mice. RESULTS: Liver weights and hepatic and fibrotic areas were decreased by 32%-52% in Cdc25A(+/-):Pkhd1(del2/del2) mice, compared with Pkhd1(del2/del2) mice. VK3 altered the cell cycle and reduced proliferation of cultured cholangiocytes by 32%-83% and decreased growth of cultured cysts by 23%-67%. In PCK rats and Pkd2(ws25/-) mice, VK3 reduced liver and kidney weights and hepato-renal cystic and fibrotic areas by 18%-34%. PM-20 decreased hepato-renal cystogenesis in Pkd2(ws25/-) mice by 15%. CONCLUSIONS: Cdc25A inhibitors block cell-cycle progression and proliferation, reduce liver and kidney weights and cyst growth in animal models of polycystic kidney disease and PLD, and might be developed as therapeutics for these diseases.


Assuntos
Cistos/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Rim/efeitos dos fármacos , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Rim Policístico Autossômico Recessivo/tratamento farmacológico , Vitamina K 3/farmacologia , Fosfatases cdc25/antagonistas & inibidores , Animais , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/enzimologia , Ductos Biliares Intra-Hepáticos/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cistos/enzimologia , Cistos/genética , Cistos/patologia , Modelos Animais de Doenças , Humanos , Rim/enzimologia , Rim/patologia , Fígado/enzimologia , Fígado/patologia , Hepatopatias/enzimologia , Hepatopatias/genética , Hepatopatias/patologia , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Rim Policístico Autossômico Recessivo/enzimologia , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Fatores de Tempo , Regulação para Cima , Fosfatases cdc25/deficiência , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
11.
Nephrol Dial Transplant ; 27(4): 1351-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22076433

RESUMO

BACKGROUND: Nephronophthisis (NPHP), the most frequent genetic cause of end-stage kidney disease in children and young adults, is characterized by a variable number of renal cysts associated with cortical tubular atrophy and interstitial fibrosis. The p38 mitogen-activated protein kinase (MAPK) pathway is an important intracellular signaling pathway involved in the production of profibrotic mediators. The relationship between p38 MAPK and renal fibrosis in NPHP2 is unknown. METHODS: We administered a selective p38 MAPK inhibitor, FR167653, in a NPHP2 mouse model (inv/inv, invΔC mice) from 3 to 6 weeks old, and the kidneys were examined at 6 weeks of age. Phosphorylation of p38 MAPK (p-p38 MAPK) protein levels, the degree of renal fibrosis, messenger RNA (mRNA) levels for extracellular matrix genes and mRNA levels for transforming growth factor in the kidneys were studied. Effect of an extracellular signal-regulated protein kinase (ERK) kinase (MEK) inhibitor on renal fibrosis was also evaluated. RESULTS: Expression of extracellular matrix genes and p-p38 MAPK were increased in the NPHP2 mouse model kidney. FR167653 successfully decreased p-p38 MAPK levels, the degree of fibrosis and extracellular matrix gene expressions. However, the FR167653 did not prevent cyst expansion, abnormal cell proliferation and acceleration of apoptosis and did not influence ERK activation. In contrast, MEK inhibition reduced both cyst expansion and fibrosis without affecting p38 MAPK activation. CONCLUSIONS: These results suggest that inhibition of p38 MAPK reduced renal fibrosis but not cyst expansion, cell proliferation and apoptosis in NPHP2 model mice. Our results suggest that p38 MAPK and ERK signaling pathways independently affect renal fibrosis in inv mutant mice.


Assuntos
Modelos Animais de Doenças , Fibrose/prevenção & controle , Nefropatias/prevenção & controle , Pirazóis/farmacologia , Piridinas/farmacologia , Fatores de Transcrição/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Apoptose , Western Blotting , Proliferação de Células , Cistos/tratamento farmacológico , Cistos/enzimologia , Cistos/prevenção & controle , Fibrose/tratamento farmacológico , Fibrose/enzimologia , Inibidores do Crescimento/farmacologia , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Camundongos , Camundongos Transgênicos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Urol Oncol ; 30(4): 463-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-20822935

RESUMO

OBJECTIVE: CA9 is proven to be a powerful marker for clear cell renal cell carcinoma. The studies on CA9 have been limited to solid renal cell carcinomas (RCC). We have conducted a study of CA9 expression in renal cystic tumors. The purpose of the present study was to extend the utility of CA9 for cystic renal tumors. MATERIALS AND METHODS: Immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) were used to detect CA9 expression in cystic renal tumors. Forty-three cystic renal tumors (22 benign and 21 malignant) were included for the immunohistochemical staining. Thirty-six patients with a cystic renal mass (20 malignant and 16 benign cystic tumors) were studied to measure CA9 level in the fluid by ELISA. Sixteen cysts (9 malignant and 7 benign cysts) were subjected both to immunohistochemistry and CA9 measurement in the fluid. RESULTS: Using immunohistochemical staining, all the benign cystic renal tumors including the 18 simple cyst and 4 benign multilocular cystic nephromas did not express CA9. All 13 cystic clear cell RCC were scored as strong staining for CA9. For 8 multilocular clear cell RCC, 7 were scored as strong staining for CA9 and the other one was negative. There was a significant difference in positive percentage (P < 0.001) between the 2 groups of malignant and benign cysts. For the 16 benign cysts, the mean concentration of CA9 in the fluid of cyst was 162 ± 133 pg/ml (median: 0 pg/ml; range: 0-2140 pg/ml). For the 20 malignant renal cystic tumors, the mean concentration of CA9 in the fluid of cyst was 2043 ± 62 pg/ml (median: 2,140 pg/ml; range: 1,112-2,140 pg/ml). There was a significant difference in mean concentration of CA9 between the two groups of malignant and benign cysts (P < 0.001). The presence or absence of CA9 expression measured by immunohistochemistry and ELISA test was concordant in 14 out of 16 cases (88%). CONCLUSIONS: Malignant cystic renal tumors expressed strongly CA9 while the benign renal cysts did not express CA9. CA9 can be detected in the fluid of malignant cystic renal tumors. CA9 is a promising molecular marker to differentiate the malignant cystic renal tumors from the benign cysts.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Anidrases Carbônicas/metabolismo , Carcinoma de Células Renais/enzimologia , Cistos/enzimologia , Doenças Renais Císticas/enzimologia , Neoplasias Renais/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anidrase Carbônica IX , Carcinoma de Células Renais/diagnóstico , Líquido Cístico/enzimologia , Cistos/diagnóstico , Diagnóstico Diferencial , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Rim/enzimologia , Rim/patologia , Doenças Renais Císticas/diagnóstico , Neoplasias Renais/diagnóstico , Masculino , Pessoa de Meia-Idade
13.
J Int Med Res ; 38(4): 1354-64, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20926008

RESUMO

Pilocytic astrocytomas are usually cystic; cyst formation within these tumours may result in increased intracranial pressure, due to the effect of their mass, and contribute to cerebral damage. Eosinophilic granular bodies (EGBs) are produced abundantly in pilocytic astrocytomas but their role in disease progression remains unknown. Immunohistochemistry studies showed EGBs to exhibit pronounced reactivity to antibodies against lysosome-associated membrane proteins (LAMP)-1 and LAMP-2, and the lysosomal enzyme cathepsin D. Both LAMP-1 and LAMP-2 showed peripheral rim and granular staining patterns. The EGBs were scattered widely across cysts and, where EGBs aggregated in clusters, were usually close to areas of fluid in the cysts. Most EGBs had nuclei either attached or close by, indicating that the EGBs may be derived from anucleated astrocytes. The results suggest that EGBs, together with other factors, may play a role in the development of cysts in pilocytic astrocytomas.


Assuntos
Astrocitoma/complicações , Catepsina D/metabolismo , Cistos/complicações , Grânulos Citoplasmáticos/enzimologia , Eosinófilos/enzimologia , Proteínas de Membrana Lisossomal/metabolismo , Adolescente , Adulto , Astrocitoma/enzimologia , Astrocitoma/patologia , Cistos/enzimologia , Cistos/patologia , Grânulos Citoplasmáticos/patologia , Feminino , Humanos , Imuno-Histoquímica , Proteína 2 de Membrana Associada ao Lisossomo , Masculino , Adulto Jovem
14.
Am J Physiol Cell Physiol ; 299(1): C21-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20457839

RESUMO

Branching morphogenesis is a fundamental process in the development of the kidney. This process gives rise to a network of ducts, which form the collecting system. Defective branching can lead to a multitude of kidney disorders including agenesis and reduced nephron number. The formation of branching tubules involves changes in cell shape, cell motility, and reorganization of the cytoskeleton. However, the exact intracellular mechanisms involved are far from understood. We have used the three-dimensional (3D) Madin-Darby canine kidney (MDCK) cell culture system to study how p21-activated kinase 1 (Pak1), which is an important regulator of the cytoskeleton, modulates branching. Our data reveal that Pak1 plays a crucial role in regulating branching morphogenesis. Expression of a dominant-negative Pak1 mutant (DN-Pak1) in MDCK cysts resulted in the spontaneous formation of extensions and branching tubules. Cellular contractility and levels of phosphorylated myosin light chain (pMLC) were increased in DN-Pak1 cells in collagen. Expression of a DN-Pak1 mutant that does not bind to PIX (DN-Pak1-DeltaPIX) failed to form extensions in collagen and did not have increased contractility. This shows that the DN-Pak1 mutant requires PIX binding to generate extensions and increased contractility in 3D culture. Furthermore, a beta1-integrin function-blocking antibody (AIIB2) inhibited the formation of branches and blocked the increased contractility in DN-Pak1 cysts. Taken together, our work shows that DN-Pak1-induced branching morphogenesis requires PIX binding and beta1-integrin signaling.


Assuntos
Cistos/enzimologia , Células Epiteliais/enzimologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Integrina beta1/metabolismo , Rim/enzimologia , Morfogênese , Quinases Ativadas por p21/metabolismo , Actinas/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Forma Celular , Colágeno Tipo I/metabolismo , Cistos/patologia , Citoesqueleto/metabolismo , Cães , Células Epiteliais/patologia , Fatores de Troca do Nucleotídeo Guanina/genética , Rim/patologia , Mutação , Cadeias Leves de Miosina/metabolismo , Fosforilação , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Fatores de Tempo , Transfecção , Quinases Ativadas por p21/genética
16.
Am J Surg Pathol ; 34(4): 556-61, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20216381

RESUMO

Cystic change in adenocarcinoma of the prostate is unusual and may be confused with benign cystic atrophy. Limited data exist on the pathologic attributes of microcystic change in malignant prostatic glands. The aim of this study was to assess histologic and immunohistologic characteristics of microcystic adenocarcinoma of the prostate. This alteration was defined as cystic dilatation and rounded expansion of the malignant gland profile, with a flat luminal cell lining layer. We identified 53 cases with microcystic change from a survey of 472 radical prostatectomy cases, for an incidence of 11.2%. The greatest diameter of the dilated cancer glands was 0.4 to 0.9 mm, with a mean diameter 10-fold greater than adjacent small malignant glands. The microcystic glands were typically adjacent to usual small acinar adenocarcinoma. Atrophic features were seen at least focally in all cases, although many microcystic adenocarcinoma epithelia exhibited a moderate amount of cytoplasm. Gleason grade 3 was the predominant grade of the adjacent nonmicrocystic malignant glands. Intraluminal crystalloids, and wispy blue intraluminal mucin were seen in all cases. Ninety-six percent of the microcystic cases showed alpha-methylacyl CoA racemase overexpression and all cases showed complete basal cell loss (using 34betaE12 and p63 antibodies) in immunohistochemistry. Microcystic adenocarcinoma of the prostate is a distinctive histomorphologic presentation of prostatic adenocarcinoma that is deceptively benign-looking at low magnifications. Detection of intraluminal crystalloids or wispy blue mucin at low magnification, immunostains for alpha-methylacyl CoA racemase, and basal cells, and a search for adjacent usual small acinar adenocarcinoma are helpful diagnostic aids. Diagnostic awareness of this growth pattern of prostatic carcinoma is important to avoid underdiagnosis of adenocarcinoma of the prostate.


Assuntos
Adenocarcinoma/patologia , Próstata/patologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Adenocarcinoma/enzimologia , Adenocarcinoma/cirurgia , Atrofia , Biomarcadores Tumorais/metabolismo , Cistos/enzimologia , Cistos/patologia , Humanos , Masculino , Próstata/enzimologia , Próstata/cirurgia , Prostatectomia , Hiperplasia Prostática/enzimologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/cirurgia , Racemases e Epimerases/metabolismo
18.
J Cell Sci ; 122(Pt 10): 1595-606, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19401335

RESUMO

The evolutionarily conserved polarity proteins PAR-3, atypical protein kinase C (aPKC) and PAR-6 critically regulate the apical membrane development required for epithelial organ development. However, the molecular mechanisms underlying their roles remain to be clarified. We demonstrate that PAR-3 knockdown in MDCK cells retards apical protein delivery to the plasma membrane, and eventually leads to mislocalized apical domain formation at intercellular regions in both two-dimensional and three-dimensional culture systems. The defects in PAR-3 knockdown cells are efficiently rescued by wild-type PAR-3, but not by a point mutant (S827/829A) that lacks the ability to interact with aPKC, indicating that formation of the PAR-3-aPKC-PAR-6 complex is essential for apical membrane development. This is in sharp contrast with tight junction maturation, which does not necessarily depend on the aPKC-PAR-3 interaction, and indicates that the two fundamental processes essential for epithelial polarity are differentially regulated by these polarity proteins. Importantly, highly depolarized cells accumulate aPKC and PAR-6, but not PAR-3, on apical protein-containing vacuoles, which become targeted to PAR-3-positive primordial cell-cell contact sites during the initial stage of the repolarization process. Therefore, formation of the PAR-3-aPKC-PAR-6 complex might be required for targeting of not only the aPKC-PAR-6 complex but also of apical protein carrier vesicles to primordial junction structures.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Moléculas de Adesão Celular/metabolismo , Comunicação Celular , Polaridade Celular , Células Epiteliais/enzimologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Vacúolos/enzimologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular , Linhagem Celular , Membrana Celular/enzimologia , Colágeno/metabolismo , Cistos/enzimologia , Cães , Géis , Humanos , Isoenzimas/genética , Camundongos , Mutação , Proteína Quinase C/genética , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Junções Íntimas/enzimologia , Fatores de Tempo , Transfecção , beta Carioferinas/metabolismo
19.
Histochem Cell Biol ; 132(2): 199-210, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19381676

RESUMO

Nephronophthisis belongs to a family of recessive cystic kidney diseases and may arise from mutations in multiple genes. In this report we have used a spontaneous mouse mutant of type 3 nephronophthisis to examine whether the doxycycline-inducible synthesis of Timp-2, a natural inhibitor of matrix metalloproteinases, can influence renal cyst growth in transgenic mice. Metalloproteinases may exert either a negative or a positive effect on the progression of cystic kidney disease, and we reasoned that this may be most effectively examined by using a natural inhibitor. Surprisingly, already the application of doxycycline, which also inhibits matrix metalloproteinases, accelerated renal cyst growth and led to increased renal fibrosis, an additional effect of Timp-2 was not detected. The positive effect of doxycycline on kidney size was not due to a non-specific "anabolic effect" but was specific for cystic kidneys because it was not observed in non-cystic kidneys. When looking for potential metabolic changes we noticed that the urine of control animals led to an increase in the calcium response of LLC-PK(1) cells, whereas the urine of doxycycline-treated mice showed the opposite effect and even antagonized the urine of control animals. Further experiments demonstrated that the urine of control animals contained a heat-labile, proteinase K-resistant substance which appears to be responsible for the induction of a calcium response in LLC-PK(1) cells. We conclude that doxycycline accelerates cyst growth possibly by the induction of a substance which lowers the intracellular calcium concentration. Our data also add a note of caution when interpreting phenotypes of animal models based upon the tet system.


Assuntos
Cistos/enzimologia , Rim/enzimologia , Rim/patologia , Doenças Renais Policísticas/enzimologia , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Animais , Cistos/induzido quimicamente , Cistos/genética , Modelos Animais de Doenças , Doxiciclina/farmacologia , Fibrose , Células HeLa , Humanos , Rim/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doenças Renais Policísticas/induzido quimicamente , Doenças Renais Policísticas/genética , Receptores de Superfície Celular/genética , Inibidor Tecidual de Metaloproteinase-2/genética
20.
EMBO J ; 27(12): 1747-57, 2008 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-18497742

RESUMO

In patients with von Hippel-Lindau (VHL) disease, renal cysts and clear cell renal cell carcinoma (ccRCC) arise from renal tubular epithelial cells containing biallelic inactivation of the VHL tumour suppressor gene. However, it is presumed that formation of renal cysts and their conversion to ccRCC involve additional genetic changes at other loci. Here, we show that cystic lesions in the kidneys of patients with VHL disease also demonstrate activation of the phosphatidylinositol-3-kinase (PI3K) pathway. Strikingly, combined conditional inactivation of Vhlh and the Pten tumour suppressor gene, which normally antagonises PI3K signalling, in the mouse kidney, elicits cyst formation after short latency, whereas inactivation of either tumour suppressor gene alone failed to produce such a phenotype. Interestingly, cells lining these cysts frequently lack a primary cilium, a microtubule-based cellular antenna important for suppression of uncontrolled kidney epithelial cell proliferation and cyst formation. Our results support a model in which the PTEN tumour suppressor protein cooperates with pVHL to suppress cyst development in the kidney.


Assuntos
Cistos/enzimologia , Cistos/patologia , Doenças Renais Císticas/enzimologia , Doenças Renais Císticas/patologia , PTEN Fosfo-Hidrolase/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Proliferação de Células , Cílios/enzimologia , Cílios/patologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Túbulos Renais Distais/enzimologia , Túbulos Renais Distais/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Mutação/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA