Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 586: 42-48, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34826699

RESUMO

Sunitinib is one of the first-line multi-tyrosine kinase inhibitors for metastatic renal cell carcinoma, and resistance to sunitinib continues to be a limiting factor for the successful treatment. As interleukin-6 (IL-6) is overexpressed in sunitinib-resistant cells, the purpose of this study was to explore the potential of IL-6 inhibition with tocilizumab, an IL-6 receptor inhibitor, to overcome resistance. In vitro, two sunitinib-resistant renal cell carcinoma cell lines (Caki-1 and SN12K1) were treated with tocilizumab. A mouse subcutaneous xenograft model was also used. Cell viability was studied by MTT assay, and apoptosis by morphology and ApopTag. Expression of IL-6, vascular endothelial growth factor (VEGF), and Bcl-2 was analyzed by qPCR. In vitro, tocilizumab induced significant cell death, and reduced the expression of IL-6, VEGF, and Bcl-2 in sunitinib-resistant cells. However, the in vitro findings could not be successfully translated in vivo, as tocilizumab did not decrease the growth of the tumors.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Renais/tratamento farmacológico , Sunitinibe/farmacologia , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos Nus , Metástase Neoplásica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cell Biol Int ; 43(6): 715-725, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31062478

RESUMO

Renal cell carcinoma (RCC) is one of the most lethal urogenital cancers and effective treatment of metastatic RCC remains an elusive target. Cell lines enable the in vitro investigation of molecular and genetic changes leading to renal carcinogenesis and are important for evaluating cellular drug response or toxicity. This study details a fast and easy protocol of establishing epithelial and fibroblast cell cultures or cell lines concurrently from renal cancer nephrectomy tissue. The protocol involves mechanical disaggregation, collagenase digestion and cell sieving for establishing epithelial cells while fibroblast cells were grown from explants. This protocol has been modified from previous published reports with additional antibiotics and washing steps added to eliminate microbial contamination from the surgical source. Cell characterisation was carried out using immunofluorescence and quantitative polymerase chain reaction. Eleven stable epithelial renal tumour cell lines of various subtypes, including rare subtypes, were established with a spontaneous immortalisation rate of 21.6% using this protocol. Eight fibroblast cell cultures grew successfully but did not achieve spontaneous immortalisation. Cells of epithelial origin expressed higher expressions of epithelial markers such as pan-cytokeratin, cytokeratin 8 and E-cadherin whereas fibroblast cells expressed high α-smooth muscle actin. Further mutational analysis is needed to evaluate the genetic or molecular characteristics of the cell lines.


Assuntos
Carcinoma de Células Renais/patologia , Cultura Primária de Células/métodos , Carcinoma de Células Renais/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Humanos , Rim/patologia , Neoplasias Renais/patologia , Nefrectomia , Células Tumorais Cultivadas
3.
Clin Exp Pharmacol Physiol ; 46(5): 456-464, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30811624

RESUMO

Glomerular crescent formation is a hallmark of rapidly progressive forms of glomerulonephritis. Thrombosis and macrophage infiltration are features of crescent formation in human and experimental kidney disease. Protease-activated receptor-2 (PAR-2) is a G-protein coupled receptor that links coagulation and inflammation. This study investigated whether pharmacological inhibition of PAR-2 can suppress glomerular crescent formation in rat nephrotoxic serum nephritis (NTN). Disease was induced in Wistar Kyoto rats by immunisation with sheep IgG followed by administration of sheep nephrotoxic serum. Rats (n = 8/group) received the PAR-2 antagonist (GB88, 10 mg/kg/p.o.), vehicle or no treatment starting 3 days before nephrotoxic serum injection and continuing until day 14. Vehicle and untreated rats developed thrombosis and macrophage infiltration in the glomerular tuft and Bowman's space in conjunction with prominent crescent formation. Activation of JNK signalling and proliferation in parietal epithelial cells was associated with crescent formation. GB88 treatment significantly reduced crescent formation with a substantial reduction in glomerular thrombosis, reduced macrophage infiltration in Bowman's space, and reduced activation of parietal epithelial cells. However, GB88 did not protect against the development of proteinuria, renal function impairment, inflammation or tubular cell damage in the NTN model. In conclusion, PAR-2 plays a specific role in glomerular crescent formation by promoting glomerular thrombosis, macrophage accumulation in Bowman's space and activation of parietal epithelial cells.


Assuntos
Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Nefrite/tratamento farmacológico , Receptor PAR-2/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Glomérulos Renais/metabolismo , Masculino , Nefrite/metabolismo , Nefrite/patologia , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Ratos , Ratos Wistar
4.
Nephrology (Carlton) ; 24(9): 983-991, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31314137

RESUMO

AIM: Protease-activated receptor 2 (PAR2) has been implicated in the development of renal inflammation and fibrosis. In particular, activation of PAR2 in cultured tubular epithelial cells induces extracellular signal-regulated kinase signalling and secretion of fibronectin, C-C Motif Chemokine Ligand 2 (CCL2) and transforming growth factor-ß1 (TGF-ß1), suggesting a role in tubulointerstitial inflammation and fibrosis. We tested this hypothesis in unilateral ureteric obstruction (UUO) in which ongoing tubular epithelial cell damage drives tubulointerstitial inflammation and fibrosis. METHODS: Unilateral ureteric obstruction surgery was performed in groups (n = 9/10) of Par2-/- and wild type (WT) littermate mice which were killed 7 days later. Non-experimental mice were controls. RESULTS: Wild type mice exhibited a 5-fold increase in Par2 messenger RNA (mRNA) levels in the UUO kidney. In situ hybridization localized Par2 mRNA expression to tubular epithelial cells in normal kidney, with a marked increase in Par2 mRNA expression by tubular cells, including damaged tubular cells, in WT UUO kidney. Tubular damage (tubular dilation, increased KIM-1 and decreased α-Klotho expression) and tubular signalling (extracellular signal-regulated kinase phosphorylation) seen in WT UUO were not altered in Par2-/- UUO. In addition, macrophage infiltration, up-regulation of M1 (NOS2) and M2 (CD206) macrophage markers, and up-regulation of pro-inflammatory molecules (tumour necrosis factor, CCL2, interleukin-36α) in WT UUO kidney were unchanged in Par2-/- UUO. Finally, the accumulation of α-SMA+ myofibroblasts, deposition of collagen IV and expression of pro-fibrotic factors (CTGF, TGF-ß1) were not different between WT and Par2-/- UUO mice. CONCLUSION: Protease-activated receptor 2 expression is substantially up-regulated in tubular epithelial cells in the obstructed kidney, but this does not contribute to the development of tubular damage, renal inflammation or fibrosis.


Assuntos
Túbulos Renais/metabolismo , Nefrite Intersticial/etiologia , Receptor PAR-2/metabolismo , Obstrução Ureteral/complicações , Animais , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Túbulos Renais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite Intersticial/genética , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Transdução de Sinais , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
5.
Am J Physiol Renal Physiol ; 314(5): F956-F968, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357409

RESUMO

Oxidative stress and mitochondrial dysfunction exacerbate acute kidney injury (AKI), but their role in any associated progress to chronic kidney disease (CKD) remains unclear. Antioxidant therapies often benefit AKI, but their benefits in CKD are controversial since clinical and preclinical investigations often conflict. Here we examined the influence of the antioxidant N-acetyl-cysteine (NAC) on oxidative stress and mitochondrial function during AKI (20-min bilateral renal ischemia plus reperfusion/IR) and progression to chronic kidney pathologies in mice. NAC (5% in diet) was given to mice 7 days prior and up to 21 days post-IR (21d-IR). NAC treatment resulted in the following: prevented proximal tubular epithelial cell apoptosis at early IR (40-min postischemia), yet enhanced interstitial cell proliferation at 21d-IR; increased transforming growth factor-ß1 expression independent of IR time; and significantly dampened nuclear factor-like 2-initiated cytoprotective signaling at early IR. In the long term, NAC enhanced cellular metabolic impairment demonstrated by increased peroxisome proliferator activator-γ serine-112 phosphorylation at 21d-IR. Intravital multiphoton microscopy revealed increased endogenous fluorescence of nicotinamide adenine dinucleotide (NADH) in cortical tubular epithelial cells during ischemia, and at 21d-IR that was not attenuated with NAC. Fluorescence lifetime imaging microscopy demonstrated persistent metabolic impairment by increased free/bound NADH in the cortex at 21d-IR that was enhanced by NAC. Increased mitochondrial dysfunction in remnant tubular cells was demonstrated at 21d-IR by tetramethylrhodamine methyl ester fluorimetry. In summary, NAC enhanced progression to CKD following AKI not only by dampening endogenous cellular antioxidant responses at time of injury but also by enhancing persistent kidney mitochondrial and metabolic dysfunction.


Assuntos
Acetilcisteína/toxicidade , Injúria Renal Aguda/complicações , Antioxidantes/toxicidade , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Insuficiência Renal Crônica/induzido quimicamente , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Metabolismo Energético/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência por Excitação Multifotônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , NAD/metabolismo , PPAR gama/metabolismo , Fosforilação , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo
6.
Toxicol Pathol ; 46(4): 449-459, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29683083

RESUMO

Indoxyl sulfate (IS) is a protein-bound uremic toxin that accumulates in patients with declining kidney function. Although generally thought of as a consequence of declining kidney function, emerging evidence demonstrates direct cytotoxic role of IS on endothelial cells and cardiomyocytes, largely through the expression of pro-inflammatory and pro-fibrotic factors. The direct toxicity of IS on human kidney proximal tubular epithelial cells (PTECs) remains a matter of debate. The current study explored the effect of IS on primary cultures of human PTECs and HK-2, an immortalized human PTEC line. Pathologically relevant concentrations of IS induced apoptosis and increased the expression of the proapoptotic molecule Bax in both cell types. IS impaired mitochondrial metabolic activity and induced cellular hypertrophy. Furthermore, statistically significant upregulation of pro-fibrotic (transforming growth factor-ß, fibronectin) and pro-inflammatory molecules (interleukin-6, interleukin-8, and tumor necrosis factor-α) in response to IS was observed. Albumin had no influence on the toxicity of IS. The results of this study suggest that IS directly induced a pro-inflammatory and pro-fibrotic phenotype in proximal tubular cells. In light of the associated apoptosis, hypertrophy, and metabolic dysfunction, this study demonstrates that IS may play a role in the progression of chronic kidney disease.


Assuntos
Apoptose/efeitos dos fármacos , Indicã/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Células Cultivadas , Humanos , Hipertrofia/patologia
7.
Int J Cancer ; 140(10): 2351-2363, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28205224

RESUMO

Metastatic renal cell carcinoma is a largely incurable disease, and existing treatments targeting angiogenesis and tyrosine kinase receptors are only partially effective. Here we reveal that MUC13, a cell surface mucin glycoprotein, is aberrantly expressed by most renal cell carcinomas, with increasing expression positively correlating with tumor grade. Importantly, we demonstrated that high MUC13 expression was a statistically significant independent predictor of poor survival in two independent cohorts, particularly in stage 1 cancers. In cultured renal cell carcinoma cells MUC13 promoted proliferation and induced the cell cycle regulator, cyclin D1, and inhibited apoptosis by inducing the anti-apoptotic proteins, BCL-xL and survivin. Silencing of MUC13 expression inhibited migration and invasion, and sensitized renal cancer cells to killing by the multi-kinase inhibitors used clinically, sorafenib and sunitinib, and reversed acquired resistance to these drugs. Furthermore, we demonstrated that MUC13 promotion of renal cancer cell growth and survival is mediated by activation of nuclear factor κB, a transcription factor known to regulate the expression of genes that play key roles in the development and progression of cancer. These results show that MUC13 has potential as a prognostic marker for aggressive early stage renal cell cancer and is a plausible target to sensitize these tumors to therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Renais/patologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Renais/patologia , Mucinas/metabolismo , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais , Western Blotting , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Técnicas Imunoenzimáticas , Indóis/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Estadiamento de Neoplasias , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Prognóstico , Pirróis/administração & dosagem , Sorafenibe , Sunitinibe , Taxa de Sobrevida , Células Tumorais Cultivadas
8.
Biochem Biophys Res Commun ; 473(1): 47-53, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26995091

RESUMO

Apoptosis repressor with caspase recruitment domain (ARC), an endogenous inhibitor of apoptosis, is upregulated in a number of human cancers, thereby conferring drug resistance and giving a rationale for the inhibition of ARC to overcome drug resistance. Our hypothesis was that ARC would be similarly upregulated and targetable for therapy in renal cell carcinoma (RCC). Expression of ARC was assessed in 85 human RCC samples and paired non-neoplastic kidney by qPCR and immunohistochemistry, as well as in four RCC cell lines by qPCR, Western immunoblot and confocal microscopy. Contrary to expectations, ARC was significantly decreased in the majority of clear cell RCC and in three (ACHN, Caki-1 and 786-0) of the four RCC cell lines compared with the HK-2 non-cancerous human proximal tubular epithelial cell line. Inhibition of ARC with shRNA in the RCC cell line (SN12K1) that had shown increased ARC expression conferred resistance to Sunitinib, and upregulated interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF). We therefore propose that decreased ARC, particularly in clear cell RCC, confers resistance to targeted therapy through restoration of tyrosine kinase-independent alternate angiogenesis pathways. Although the results are contrary to expectations from other cancer studies, they were confirmed here with multiple analytical methods. We believe the highly heterogeneous nature of cancers like RCC predicate that expression patterns of molecules must be interpreted in relation to respective matched non-neoplastic regions. In the current study, this procedure indicated that ARC is decreased in RCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Proteínas do Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos , Indóis/uso terapêutico , Neoplasias Renais/metabolismo , Neovascularização Patológica , Proteínas do Tecido Nervoso/metabolismo , Pirróis/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/química , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Nephrology (Carlton) ; 21(3): 170-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26239363

RESUMO

In the last decade, chronic kidney disease (CKD), defined as reduced renal function (glomerular filtration rate (GFR) < 60 mL/min per 1.73 m(2) ) and/or evidence of kidney damage (typically manifested as albuminuria) for at least 3 months, has become one of the fastest-growing public health concerns worldwide. CKD is characterized by reduced clearance and increased serum accumulation of metabolic waste products (uremic retention solutes). At least 152 uremic retention solutes have been reported. This review focuses on indoxyl sulphate (IS), a protein-bound, tryptophan-derived metabolite that is generated by intestinal micro-organisms (microbiota). Animal studies have demonstrated an association between IS accumulation and increased fibrosis, and oxidative stress. This has been mirrored by in vitro studies, many of which report cytotoxic effects in kidney proximal tubular cells following IS exposure. Clinical studies have associated IS accumulation with deleterious effects, such as kidney functional decline and adverse cardiovascular events, although causality has not been conclusively established. The aims of this review are to: (i) establish factors associated with increased serum accumulation of IS; (ii) report effects of IS accumulation in clinical studies; (iii) critique the reported effects of IS in the kidney, when administered both in vivo and in vitro; and (iv) summarize both established and hypothetical therapeutic options for reducing serum IS or antagonizing its reported downstream effects in the kidney.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal , Indicã/sangue , Rim/metabolismo , Insuficiência Renal Crônica/sangue , Animais , Dieta com Restrição de Proteínas , Fibras na Dieta/administração & dosagem , Disbiose , Taxa de Filtração Glomerular , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Probióticos , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/terapia , Regulação para Cima , Agentes Urológicos/uso terapêutico
10.
Biochem Cell Biol ; 93(1): 63-73, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25453190

RESUMO

A growing body of evidence demonstrates the involvement of protein modification with O-linked ß-N-acetylglucosamine (O-GlcNAc) in the stress response and its beneficial effects on cell survival. Here we investigated protein O-GlcNAcylation in skeletal muscle cells exposed to oxidative stress and the crosstalk with endogenous antioxidant system. The study focused on antioxidant enzymes superoxide dismutase 2 (SOD2), catalase (CAT), and glutathione peroxidase 1 (GPX1), and transcriptional regulators proliferator-activated receptor gamma coactivator 1-α (PGC-1α) and forkhead box protein O1 (FOXO1), which play important roles in oxidative stress response and are known to be O-GlcNAc-modified. C2C12 myoblasts were subjected to 24 h incubation with different reagents, including hydrogen peroxide, diethyl maleate, high glucose, and glucosamine, and the inhibitors of O-GlcNAc cycling enzymes. Surprisingly, O-GlcNAc levels were significantly increased only with glucosamine, whilst other treatments showed no effect. Significant changes at the mRNA level were observed with concomitant upregulation of the genes for O-GlcNAc enzymes and stress-related proteins with oxidizing agents and downregulation of these genes with agents promoting O-GlcNAcylation. Our findings suggest a role of O-GlcNAc in the stress response and indicate an inhibitory mechanism controlling O-GlcNAc levels in the muscle cells. This could represent an important homeostatic regulation of the cellular defense system.


Assuntos
Acetilglucosamina/farmacologia , Mioblastos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes , Catalase/genética , Linhagem Celular , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Glutationa Peroxidase/genética , Proteínas de Choque Térmico , Camundongos , Oxidantes , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Processamento de Proteína Pós-Traducional , Superóxido Dismutase/genética , Fatores de Transcrição/genética , Glutationa Peroxidase GPX1
11.
Environ Toxicol ; 30(12): 1423-33, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24942245

RESUMO

Thimerosal is an ethyl mercury-containing compound used mainly in vaccines as a bactericide. Although the kidney is a key target for mercury toxicity, thimerosal nephrotoxicity has not received the same attention as other mercury species. The aim of this study was to determine the potential cytotoxic mechanisms of thimerosal on human kidney cells. Human kidney proximal tubular epithelial (HK2) cells were exposed for 24 h to thimerosal (0-2 µM), and assessed for cell viability, apoptosis, and cell proliferation; expression of proteins Bax, nuclear factor-κB subunits, and transforming growth factor-ß1 (TGFß1); mitochondrial health (JC-1, MitoTracker Red CMXRos); and fibronectin levels (enzyme-linked immunosorbent assay). Thimerosal diminished HK2 cell viability and mitosis, promoted apoptosis, impaired the mitochondrial permeability transition, enhanced Bax and TGFß1 expression, and augmented fibronectin secretion. This is the first report about kidney cell death and pro-fibrotic mechanisms promoted by thimerosal. Collectively, these in vitro results demonstrate that (1) thimerosal induces kidney epithelial cell apoptosis via upregulating Bax and the mitochondrial apoptotic pathway, and (2) thimerosal is a potential pro-fibrotic agent in human kidney cells. We suggest that new evidence on toxicity as well as continuous surveillance in terms of fibrogenesis is required concerning thimerosal use.


Assuntos
Apoptose/efeitos dos fármacos , Timerosal/toxicidade , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibronectinas/análise , Fibrose , Humanos , Túbulos Renais/citologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitose/efeitos dos fármacos , NF-kappa B/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína X Associada a bcl-2/metabolismo
12.
Am J Physiol Renal Physiol ; 307(7): F814-22, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25122050

RESUMO

The mechanism(s) underlying renoprotection by peroxisome proliferator-activated receptor (PPAR)-γ agonists in diabetic and nondiabetic kidney disease are not well understood. Mitochondrial dysfunction and oxidative stress contribute to kidney disease. PPAR-γ upregulates proteins required for mitochondrial biogenesis. Our aim was to determine whether PPAR-γ has a role in protecting the kidney proximal tubular epithelium (PTE) against mitochondrial destabilisation and oxidative stress. HK-2 PTE cells were subjected to oxidative stress (0.2-1.0 mM H2O2) for 2 and 18 h and compared with untreated cells for apoptosis, mitosis (morphology/biomarkers), cell viability (MTT), superoxide (dihydroethidium), mitochondrial function (MitoTracker red and JC-1), ATP (luminescence), and mitochondrial ultrastructure. PPAR-γ, phospho-PPAR-γ, PPAR-γ coactivator (PGC)-1α, Parkin (Park2), p62, and light chain (LC)3ß were investigated using Western blots. PPAR-γ was modulated using the agonists rosiglitazone, pioglitazone, and troglitazone. Mitochondrial destabilization increased with H2O2concentration, ATP decreased (2 and 18 h; P < 0.05), Mitotracker red and JC-1 fluorescence indicated loss of mitochondrial membrane potential, and superoxide increased (18 h, P < 0.05). Electron microscopy indicated sparse mitochondria, with disrupted cristae. Mitophagy was evident at 2 h (Park2 and LC3ß increased; p62 decreased). Impaired mitophagy was indicated by p62 accumulation at 18 h (P < 0.05). PPAR-γ expression decreased, phospho-PPAR-γ increased, and PGC-1α decreased (2 h), indicating aberrant PPAR-γ activation and reduced mitochondrial biogenesis. Cell viability decreased (2 and 18 h, P < 0.05). PPAR-γ agonists promoted further apoptosis. In summary, oxidative stress promoted mitochondrial destabilisation in kidney PTE, in association with increased PPAR-γ phosphorylation. PPAR-γ agonists failed to protect PTE. Despite positive effects in other tissues, PPAR-γ activation appears to be detrimental to kidney PTE health when oxidative stress induces damage.


Assuntos
Apoptose , Túbulos Renais Proximais/metabolismo , Renovação Mitocondrial , Estresse Oxidativo , PPAR gama/metabolismo , Linhagem Celular , Humanos , Urotélio/metabolismo
13.
Biochem Biophys Res Commun ; 444(3): 332-7, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24462876

RESUMO

The use of recombinant human erythropoietin (rhEPO) to promote repair and minimize cardiac hypertrophy after myocardial infarction has had disappointing outcomes in clinical trials. We hypothesized that the beneficial non-hematopoietic effects of rhEPO against cardiac hypertrophy could be offset by the molecular changes initiated by rhEPO itself, leading to rhEPO resistance or maladaptive hypertrophy. This hypothesis was investigated using an isoproterenol-induced model of myocardial infarct and cardiac remodelling with emphasis on hypertrophy. In h9c2 cardiomyocytes, rhEPO decreased isoproterenol-induced hypertrophy, and the expression of the pro-fibrotic factors fibronectin, alpha smooth muscle actin and transforming growth factor beta-1 (TGF-ß1). In contrast, by itself, rhEPO increased the expression of fibronectin and TGF-ß1. Exogenous TGF-ß1 induced a significant increase in hypertrophy, which was further potentiated by rhEPO. Exogenous fibronectin not only induced hypertrophy of cardiomyocytes, but also conferred resistance to rhEPO treatment. Based on these findings we propose that the outcome of rhEPO treatment for myocardial infarction is determined by the baseline concentrations of fibronectin and TGF-ß1. If endogenous fibronectin or TGF-ß levels are above a certain threshold, they could cause resistance to rhEPO therapy and enhancement of cardiac hypertrophy, respectively, leading to maladaptive hypertrophy.


Assuntos
Cardiomegalia/fisiopatologia , Eritropoetina/farmacologia , Fibronectinas/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/tratamento farmacológico , Linhagem Celular , Isoproterenol/toxicidade , Ratos , Proteínas Recombinantes/farmacologia
14.
Am J Pathol ; 180(4): 1702-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22322296

RESUMO

Caveolin-1 is an essential structural protein of caveolae, specialized plasma membrane organelles highly abundant in endothelial cells, where they regulate multiple functions including angiogenesis. Caveolin-1 exerts a tonic inhibition of endothelial nitric oxide synthase (eNOS) activity. Accordingly, caveolin-1 gene-disrupted mice have enhanced eNOS activity as well as increased systemic nitric oxide (NO) levels. We hypothesized that excess eNOS activity, secondary to caveolin deficiency, would mediate the decreased angiogenesis observed in caveolin-1 gene-disrupted mice. We tested tumor angiogenesis in mice lacking either one or both proteins, using in vitro, ex vivo, and in vivo assays. We show that endothelial cell migration, tube formation, cell sprouting from aortic rings, tumor growth, and angiogenesis are all significantly impaired in both caveolin-1-null and eNOS-null mice. We further show that these parameters were either partially or fully restored in double knockout mice that lack both caveolin-1 and eNOS. Furthermore, the effects of genetic ablation of eNOS are mimicked by the administration of the NOS inhibitor N-nitro-L-arginine methyl ester hydrochloride (L-NAME), including the reversal of the caveolin-1-null mouse angiogenic phenotype. This study is the first to demonstrate the detrimental effects of unregulated eNOS activity on angiogenesis, and shows that impaired tumor angiogenesis in caveolin-1-null mice is, at least in part, the result of enhanced eNOS activity.


Assuntos
Caveolina 1/deficiência , Neovascularização Patológica/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Animais , Caveolina 1/genética , Caveolina 1/fisiologia , Células Cultivadas , Quimiotaxia/fisiologia , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/prevenção & controle , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Inibidores Enzimáticos/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , NG-Nitroarginina Metil Éster/uso terapêutico , Transplante de Neoplasias , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
BMC Cancer ; 13: 14, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23305401

RESUMO

One of the molecules regulated by the transcription factor, hypoxia inducible factor (HIF), is the hypoxia-responsive hematopoietic factor, erythropoietin (EPO). This may have relevance to the development of renal cell carcinoma (RCC), where mutations of the von Hippel-Lindau (VHL) gene are major risk factors for the development of familial and sporadic RCC. VHL mutations up-regulate and stabilize HIF, which in turn activates many downstream molecules, including EPO, that are known to promote angiogenesis, drug resistance, proliferation and progression of solid tumours. HIFs typically respond to hypoxic cellular environment. While the hypoxic microenvironment plays a critical role in the development and progression of tumours in general, it is of special significance in the case of RCC because of the link between VHL, HIF and EPO. EPO and its receptor, EPOR, are expressed in many cancers, including RCC. This limits the use of recombinant human EPO (rhEPO) to treat anaemia in cancer patients, because the rhEPO may be stimulatory to the cancer. EPO may also stimulate epithelial-mesenchymal transition (EMT) in RCC, and pathological EMT has a key role in cancer progression. In this mini review, we summarize the current knowledge of the role of EPO in RCC. The available data, either for or against the use of EPO in RCC patients, are equivocal and insufficient to draw a definitive conclusion.


Assuntos
Carcinoma de Células Renais/metabolismo , Eritropoetina/metabolismo , Neoplasias Renais/metabolismo , Anemia/sangue , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Carcinoma de Células Renais/sangue , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Eritropoetina/efeitos adversos , Hematínicos/efeitos adversos , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/sangue , Neoplasias Renais/genética , Neoplasias Renais/patologia , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/efeitos adversos , Fatores de Risco , Transdução de Sinais , Microambiente Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
16.
PLoS One ; 16(3): e0248983, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33765016

RESUMO

Expression of the protease sensing receptor, protease activated receptor-2 (PAR2), is elevated in a variety of cancers and has been promoted as a potential therapeutic target. With the development of potent antagonists for this receptor, we hypothesised that they could be used to treat renal cell carcinoma (RCC). The expression of PAR2 was, therefore, examined in human RCC tissues and selected RCC cell lines. Histologically confirmed cases of RCC, together with paired non-involved kidney tissue, were used to produce a tissue microarray (TMA) and to extract total tissue RNA. Immunohistochemistry and qPCR were then used to assess PAR2 expression. In culture, RCC cell lines versus primary human kidney tubular epithelial cells (HTEC) were used to assess PAR2 expression by qPCR, immunocytochemistry and an intracellular calcium mobilization assay. The TMA revealed an 85% decrease in PAR2 expression in tumour tissue compared with normal kidney tissue. Likewise, qPCR showed a striking reduction in PAR2 mRNA in RCC compared with normal kidney. All RCC cell lines showed lower levels of PAR2 expression than HTEC. In conclusion, we found that PAR2 was reduced in RCC compared with normal kidney and is unlikely to be a target of interest in the treatment of this type of cancer.


Assuntos
Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Receptor PAR-2/metabolismo , Biópsia , Cálcio/metabolismo , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , Túbulos Renais/patologia , Receptor PAR-2/genética
17.
Apoptosis ; 15(4): 412-25, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19856104

RESUMO

One of the impeding factors in the effective treatment of metastatic renal cell carcinoma (RCC) is their intrinsic and acquired resistance to chemotherapeutics. Many studies have shown that drug resistance, at least in part, is mediated by the upregulation of anti-apoptotic (Bcl-2) and multidrug resistance molecules (MDR-1 and MRP-1) by the transcription factor nuclear factor kappa B (NF-kappaB). Combining NF-kappaB inhibitors with conventional chemotherapeutics could overcome resistance of cancer cells. In this study, we examined the synergistic effect of pyrrolidine dithiocarbamate (PDTC), a NF-kappaB inhibitor, and cisplatin, on two human metastatic RCC cell lines ACHN and SN12K1. Individual non-toxic concentrations of PDTC and cisplatin, when combined, synergistically induced a significant increase in apoptosis of the two RCC cell lines. In ACHN cells, the groups with nuclear translocation of NF-kappaB showed resistance to apoptosis, but in SN12K1 cells, the groups with NF-kappaB translocation were susceptible to apoptosis. The combination treatment significantly decreased the transcription activity of all NF-kappaB subunits in both cell lines. Anti-apoptotic proteins Bcl-2 and Bcl-(XL) were significantly decreased in the combination therapy group of both cell lines, but MDR-1 was decreased only in the ACHN cells. No changes in MRP-1 were observed in any of the treatment groups. The results demonstrate the potential of PDTC to be an adjunct therapeutic agent. The major mechanism of the synergistic effect appears to be mediated by the inhibition of transcription activity of NF-kappaB rather than its expression, and the resultant decrease in the anti-apoptotic proteins Bcl-2 and Bcl-(XL).


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Cisplatino/uso terapêutico , Neoplasias Renais/tratamento farmacológico , NF-kappa B/genética , Pirrolidinas/uso terapêutico , Tiocarbamatos/uso terapêutico , Transcrição Gênica , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Renais/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Subunidades Proteicas/genética , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteína bcl-X/metabolismo
18.
Nephrol Dial Transplant ; 25(5): 1462-74, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20037166

RESUMO

BACKGROUND: Renal cell carcinoma (RCC) is a highly metastatic and lethal disease with few efficacious treatments. Many studies have shown that the ubiquitous transcription factor nuclear factor kappa B (NF-kappaB) plays a key role in the development and progression of many cancers including RCC. The aim of this investigation was to evaluate the anti-cancer effect of pyrrolidine dithiocarbamate (PDTC), a NF-kappaB inhibitor, in a murine xenograft model of RCC. METHODS: The metastatic human RCC cell line, SN12K1, was inoculated into the left kidneys of severe combined immunodeficiency mice and the effect of semi-continuous PDTC treatment (50 mg/kg) on RCC growth analysed 5 weeks later. The analyses carried out in three groups (no treatment, RCC alone and RCC + PDTC) at 5 weeks were: renal weight, protein expression by immunohistochemistry and Western immunoblot, apoptosis (TdT-mediated nick end labelling and morphology) and mitosis (morphology). RESULTS: PDTC significantly decreased RCC growth and the expression of NF-kappaB subunits (p50, p52, c-Rel and RelB), upstream IKK-beta and IKK-gamma, but did not induce any changes in the expression of IkappaB-alpha and IkappaB-beta. RCC growth was associated with a significant decrease in the expression of the anti-apoptotic proteins Bcl-2 and Bcl-(XL) and increase in pro-apoptotic Bax, all of which were reversed by PDTC. Cell proliferation was significantly reduced by PDTC. CONCLUSION: The results demonstrate the potential anti-cancer benefits of treating NF-kappaB positive RCCs with NF-kappaB inhibitors like PDTC.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Pirrolidinas/uso terapêutico , Tiocarbamatos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Humanos , Quinase I-kappa B/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos SCID , NF-kappa B/genética , Proteínas Proto-Oncogênicas c-bcl-2/análise , Redução de Peso/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína bcl-X/análise
19.
Angiogenesis ; 12(4): 365-79, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19882112

RESUMO

Renal cell carcinomas (RCC) are a heterogeneous group of cancers that often include angiogenesis as a key clinical and pathological hallmark. As the transcription factor nuclear factor kappa B (NF-kappaB) has been identified as a key promoter of angiogenesis, NF-kappaB inhibitors could serve as potential anti-angiogenic agents. In this study, we tested the anti-angiogenic properties of pyrrolidine dithiocarbamate (PDTC), a NF-kappaB inhibitor, using established in vitro and ex vivo assays in human umbilical vein endothelial cells (HUVEC) and human metastatic RCC cell lines (ACHN and SN12K1). In vitro, PDTC inhibited proliferation, capillary tube formation, invasion and trans-differentiation of HUVEC. Ex vivo, PDTC blocked vessel sprouting from aortic explants and disrupted the integrity of pre-formed vessels. PDTC also inhibited the adhesion of HUVEC and RCC cells to substratum and inhibited their invasion. PDTC inhibited RCC-induced proliferation of HUVEC. Protein microarray demonstrated heterogenic actions in each cell line: in HUVEC, epidermal growth factor was significantly decreased; in ACHN, basic fibroblast growth factor, growth related oncogene, interleukin-6, RANTES and monocyte chemoattractant protein-1 (MCP-1) were significantly decreased; and in SN12K1, MCP-1 was upregulated. PDTC increased the expression of the pro-angiogenic protein interleukin-8 in both RCC cell lines. Expression of vascular endothelial growth factor (VEGF) was not significantly altered, and exogenous VEGF had a neutral effect on in vitro angiogenesis of HUVEC. Collectively, these data suggest that PDTC has some anti-angiogenic properties, which may limit development and progression of RCC.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Prolina/análogos & derivados , Tiocarbamatos/farmacologia , Sequência de Aminoácidos , Animais , Aorta/citologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/secundário , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Endoteliais/citologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Renais/patologia , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Técnicas de Cultura de Órgãos , Peptídeos/farmacologia , Prolina/farmacologia , Ratos , Ratos Sprague-Dawley , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
20.
Transl Androl Urol ; 8(Suppl 2): S123-S137, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31236330

RESUMO

BACKGROUND: Differentiation of chromophobe renal cell carcinoma (chRCC) from benign renal oncocytoma (RO) can be challenging especially when there are overlapping histological and morphological features. In this study we have investigated immunohistochemical biomarkers (cytokeratin 7/CK7, Caveolin-1/Cav-1 and S100 calcium-binding protein A1/S100A1) to aid in this difficult differentiation and attempted to validate their use in human renal tumour tissue to assess their discriminatory ability, particularly for chRCC and RO, in an Australian cohort of patients. METHODS: Retrospective study was carried out of archived formalin-fixed paraffin-embedded renal tumours from tumour nephrectomy specimens of 75 patients: 30 chRCC, 15 RO and 30 clear cell RCC (ccRCC). Sections were cut and immunostained with specific polyclonal antibodies of CK7, Cav-1 and S100A1. Morphometry was used to determine expression patterns of the biomarkers using Aperio ImageScope. Results were assessed with student t-test and ANOVA with significance at P<0.05. RESULTS: From this cohort, male-to-female ratio was 1.9:1. Median age was 64 (45-88 years) and median tumour size was 3.8 cm (range, 1.2-18 cm). There were 47 (62.7%) T1, 7 T2, 20 T3 and one T4 stage of RCC; with 2 patients presenting with M1 stage. There was significantly higher CK7 expression in chRCC compared to RO (P=0.03), and chRCC also had a different staining pattern and higher expression of Cav-1 compared to RO. There was higher expression of S100A1 in RO compared to chRCC. CONCLUSIONS: Immunohistochemical staining and standard morphometry of CK7, Cav-1 and S100A1 can aid in the differentiation of chRCC and RO. This may guide clinicians in management of patients when faced with difficult diagnostic histological distinction between the two tumour subtypes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA