Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1156120, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37900138

RESUMO

Introduction: Prostate cancer (PCa) presents a significant health challenge in men, with a substantial number of deaths attributed to metastatic castration resistant PCa (mCRPC). Moreover, African American men experience disproportionately high mortality rates due to PCa. This study delves into the pivotal role of SPDEF, a prostate specific Ets transcription factor, and its regulation by DNA methylation in the context of PCa progression. Methods: We performed Epigenetic reprogramming using daily treatment with non-toxic dose of 5Aza-2-deoxycytidine (5Aza-dC) for two weeks to assess its impact on PDEF expression in prostate cancer cells. Next, we conducted functional studies on reprogrammed cells, including cell migration (wound-healing assay), invasion (Boyden-Chamber test), and proliferation (MTT assay) to comprehensively evaluate the consequences of altered PDEF expression. We used bisulfite sequencing (BSP) to examine DNA methylation at SPDEF promoter. Simultaneously, we utilized siRNA-mediated targeting of key DNMTs (DNMT1, DNMT3A, and DNMT3B) to elucidate their specific role in regulating PDEF. We measured mRNA and protein expressions using qRT-PCR and immune-blotting techniques, respectively. Results: In this report, we observed that: a) there is a gradual decrease in SPDEF expression with a concomitant increase in methylated CpG sites within the SPDEF gene during prostate cancer progression from lower to higher Gleason grade; b) Expression of DNMT's (DNMT1, 3a and 3b) is increased during prostate cancer progression, and there is an inverse correlation between SPDEF and DNMT expression; c) SPDEF levels are decreased in RC77/T, a line of PCa cells from African American origin similar to PC3 and DU145 cells (CRPC cells), as compared to LNCaP cells , a line of androgen dependent cells,; d) the 5' CpG island of SPDEF gene are hypermethylated in SPDEF-negative CRPC ( PC3, DU145 and RC77/T) cell lines but the same regions are hypomethylated in SPDEF-positive castrate sensitive (LNCaP) cell line ; (e) expression of SPDEF in PCa cells lacking SPDEF decreases cell migration and invasion, but has no significant effect on cell proliferation, and; (f) treatment with the demethylating agent, 5-aza-2'-deoxycytidine, or silencing of the DNMT's by siRNA, partially restores SPDEF expression in SPDEF-negative PCa cell lines, and decreases cell migration and invasion. Discussion: These results indicate hypermethylation is a prevalent mechanism for decreasing SPDEF expression during prostate cancer progression. The data demonstrate that loss of SPDEF expression in prostate cancer cells, a critical step in cellular plasticity, results from a potentially reversible process of aberrant DNA methylation. These studies suggest DMNT activity as a potential therapeutic vulnerability that can be exploited for limiting cellular plasticity, tumor progression, and therapy resistance in prostate cancer.


Assuntos
Metilação de DNA , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Neoplasias de Próstata Resistentes à Castração/genética , Linhagem Celular Tumoral , Ilhas de CpG/genética , Decitabina , RNA Interferente Pequeno , Proteínas Proto-Oncogênicas c-ets/genética
2.
Cureus ; 13(3): e13831, 2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33854851

RESUMO

Introduction Obesity, defined as a condition of excessive fat accumulation in adipose tissue, is a global epidemic implicated in a myriad of processes deleterious to human health. It has become one of the leading impediments to public health globally. The study of obesity necessitates adipocyte models, which commonly employ a medium enriched with adipogenic hormones and fetal bovine serum (FBS) to culture terminal adipocytes. In the current study, we developed a novel protocol for serum-free differentiation of 3T3-L1 and ST2 pre-adipocytes using media enriched with free fatty acids (FFA) and bovine serum albumin (BSA). Differentiation was characterized by measuring FFA uptake and changes in expression of adipogenic genes. The novel protocol was also compared against the existing serum-inclusive method. Methods The National Institutes of Health (NIH)-3T3-L1 and ST2 pre-adipocyte cells were maintained in Dulbecco's Modified Eagle Medium (DMEM) containing 10% calf serum and 1% penicillin-streptomycin and Roswell Park Memorial Institute Medium (RPMI) with 10% FBS and 1% penicillin-streptomycin mixture, respectively, at 37℃, 5% CO2 in a humidified atmosphere. Differentiation was induced using a mixture of 0.25 µM dexamethasone, 0.5 mM 3-isobutyl-1-methylxanthine (IBMX), 10 µg/mL insulin, or 1% insulin-transferrin-selenium (ITS). Cells were cultured in serum-free media containing DMEM with BSA (2.5%) and lipid mixture 1 (LM1 1%) as well as serum-inclusive media enriched with 10% FBS. Total RNA was extracted, and quantitative reverse transcription-polymerase chain reaction (RT-PCR) was performed using delta-delta Ct method, also known as the 2-∆∆Ct  method. Ribosomal protein, large, P0 (RPLP0) was used as a house-keeping gene for quantitation of relative expressions. Results We observed an increase in fatty acid accumulation relative to controls using Oil Red O neutral lipid staining and spectrophotometry. This result was consistent with the effects of the serum-inclusive method. Differentiation was further confirmed by increased gene expression of adipogenic transcription factors - peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (C/EBPα); adipogenic genes - fatty acid-binding protein 4 (FABP4/aP2) and fatty acid translocase (FAT/CD36); and the lipogenic gene - perilipin by using quantitative RT-PCR. Conclusion Our data suggest that serum-free differentiation can significantly enhance the free fatty acid accumulation as well as adipogenic gene expression in both NIH-3T3-L1 and ST2 pre-adipocyte cells. Given the shortcomings of FBS, this method may provide advantages to the serum-inclusive protocols described previously.

3.
Hum Cell ; 32(3): 240-250, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30875077

RESUMO

Intestinal epithelial differentiation may be stimulated by diverse pathways including luminal short-chain fatty acids and repetitive mechanical deformation engendered by villous motility and peristalsis. Schlafen 12 (SLFN12) is a cytosolic protein that stimulates sucrase-isomaltase (SI) expression. We hypothesized that two disparate differentiating stimuli, butyrate and repetitive deformation, would each stimulate SLFN12 expression in human Caco-2 intestinal epithelial cells and that increased SLFN12 expression would contribute to the differentiating activity of the human Caco-2 intestinal epithelial cells. We stimulated Caco-2 cells with 1-2 mM butyrate or repetitive mechanical deformation at 10 cycles/min at an average 10% strain, and measured SLFN12 and SI expression by qRT-PCR. Sodium butyrate enhanced SLFN12 expression at both 1 mM and 2 mM although SI expression was only significantly increased at 2 mM. Repetitive deformation induced by cyclic mechanical strain also significantly increased both SLFN12 and SI gene expression. Reducing SLFN12 by siRNA decreased basal, deformation-stimulated, and butyrate-stimulated SLFN12 levels, compared to control cells treated with non-targeting siRNA, although both deformation and butyrate were still able to stimulate SLFN12 expression in siRNA-treated cells compared to control cells treated with the same siRNA. This attenuation of the increase in SLFN12 expression in response to mechanical strain or butyrate was accompanied by parallel attenuation of SI expression. Butyrate stimulated SI-promoter activity, and reducing SLFN12 by siRNA attenuated butyrate-induced SI-promoter activity. These data suggest that SLFN12 mediates at least in part the stimulation by both butyrate and repetitive mechanical deformation of sucrase-isomaltase, a late stage differentiation marker in human intestinal epithelial cells.


Assuntos
Ácido Butírico/farmacologia , Proteínas de Transporte/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Epiteliais/fisiologia , Motilidade Gastrointestinal/fisiologia , Intestinos/citologia , Peristaltismo/fisiologia , Células CACO-2 , Carbidopa , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Expressão Gênica/efeitos dos fármacos , Humanos , Levodopa/análogos & derivados , Complexo Sacarase-Isomaltase/metabolismo
4.
Cell Physiol Biochem ; 48(3): 1274-1290, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30045019

RESUMO

BACKGROUND/AIMS: Human enterocytic differentiation is altered during development, fasting, adaptation, and bariatric surgery, but its intracellular control remains unclear. We hypothesized that Schlafen 12 (SLFN12) regulates enterocyte differentiation. METHODS: We used laser capture dissection of epithelium, qRT-PCR, and immunohistochemistry to evaluate SLFN12 expression in biopsies of control and fasting human duodenal mucosa, and viral overexpression and siRNA to trace the SLFN12 pathway in human Caco-2 and HIEC6 intestinal epithelial cells. RESULTS: Fasting human duodenal mucosa expressed less SLFN12 mRNA and protein, accompanied by decreases in enterocytic markers like sucrase-isomaltase. SLFN12 overexpression increased Caco-2 sucrase-isomaltase promoter activity, mRNA, and protein independently of proliferation, and activated the SLFN12 putative promoter. SLFN12 coprecipitated Serpin B12 (SERPB12). An inactivating SLFN12 point mutation prevented both SERPB12 binding and sucrase-isomaltase induction. SERPB12 overexpression also induced sucrase-isomaltase, while reducing SERPB12 prevented the SLFN12 effect on sucrase-isomaltase. Sucrase-isomaltase induction by both SLFN12 and SERPB12 was attenuated by reducing UCHL5 or USP14, and blocked by reducing both. SERPB12 stimulated USP14 but not UCHL5 activity. SERPB12 coprecipitated USP14 but not UCHL5. Moreover, SLFN12 increased protein levels of the sucrase-isomaltase-promoter-binding transcription factor cdx2 without altering Cdx2 mRNA. This was prevented by reducing UCHL5 and USP14. We further validated this pathway in vitro and in vivo. SLFN12 or SERPB12 overexpression induced sucrase-isomaltase in human non-malignant HIEC-6 enterocytes. CONCLUSIONS: SLFN12 regulates human enterocytic differentiation by a pathway involving SERPB12, the deubiquitylases, and Cdx2. This pathway may be targeted to manipulate human enterocytic differentiation in mucosal atrophy, short gut or obesity.


Assuntos
Diferenciação Celular , Enzimas Desubiquitinantes/metabolismo , Enterócitos/citologia , Mapas de Interação de Proteínas , Proteínas/metabolismo , Serpinas/metabolismo , Células CACO-2 , Células Cultivadas , Enterócitos/metabolismo , Jejum , Humanos
5.
Oncotarget ; 8(44): 76574-76586, 2017 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-29100333

RESUMO

BACKGROUND: The codon 72 polymorphism in p53 has been implicated in colorectal cancer (CRC) risk, prognosis and CRC health disparities. We examined the functional consequence of this polymorphism in CRC. EXPERIMENTAL DESIGN: Plasmids (pCMV6) that express different phenotypes of p53 [p53 wild type (wt) at codon 72 (R72wt), R72wt with mutation at codon 273 cysteine (R72273Cys), p53 mutation at codon 72 (P72wt) and P72wt with mutation at codon 273 (P72273Cys)] were constructed. The CRC cell line Caco2, which does not express p53 for in vitro studies, was used as host. CRC xenografts were established in severe combined immunodeficient (SCID) mice using established cell lines. CRC surgical specimens, corresponding normal colon, and tumor xenografts were sequenced for codon 72 polymorphism of p53. Proteins signaling mechanisms were evaluated to assess the functional consequence of P72 phenotype of p53. RESULTS: This study demonstrated a significantly increased survival of cells expressing P72wt, mutant phenotype, versus R72wt phenotype. WB analyses revealed that P72wt induced activation of p38 and RAF/MEK/ extracellular signal-regulated kinase (ERK) MAP kinases. Activation of CREB was found to be higher in tumors that exhibit P72 phenotype. Metastatic lesions of CRC expressed more phospho-CREB than non-metastatic lesions. The expression of P72wt promoted CRC metastasis. CONCLUSIONS: P72 contributes to the aggressiveness of CRC. Because P72 is over-expressed in CRC, specifically in African-American patients, this suggests a role for P72 in cancer health disparities. This work was supported by NIH/NCI Workforce Diversity Grant R21-CA171251 & U54CA118948.

6.
J Nanosci Nanotechnol ; 17(1): 175-85, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29617099

RESUMO

Triple-Negative Breast Cancer (TNBC) has a poor prognosis due to lack of targeted therapy. Doxorubicin (DOX) has failed for multiple reasons, including development of multi-drug resistance, induction of inflammation (IL-6 secretion) and long-term toxicities. DOX is also known to have off target proteasomal activation, justifying the concept of combining it with a proteasomal inhibitor. Our study investigated the therapeutic potential of an irreversible proteasome inhibitor carfilzomib (CARF) alone or in combination with DOX in two TNBC cell lines (MDA-MB-231 and MDA-MB-468). CARF was as effective in inhibiting mitosis in vitro for both cell lines in comparison to DOX alone. CARF performed similar to DOX in inhibiting apoptosis but had better results in reducing proliferation. Further studies in MDA-MB-231 cells demonstrated that CARF also inhibited pro-inflammatory IL-6 secretion and NF κB transcriptional activity while DOX stimulated both IL-6 and NF kappa-B activity. The reduction of IL-6 while using CARF highlights its therapeutic potential and ability to enhance current clinical drug regimens. Furthermore, exogenous administration of IL-6 potentiated NF Kappa B transcriptional activity, pSTAT3 (Tyr705) and JAK inflammatory signaling as well as cell proliferation in CARF- or DOX-treated MDA-MB-231 cells. In vivo, CARF treatment resulted in reduced serum IL-6 compared to treatment with DOX in female SCID-NOD mice with MDA-MB-231 cell tumor. A combinational approach using DOX and CARF presents a clinical potential for better efficacy, reduced proliferation, apoptosis, anti-angiogenesis, and less cardiac dysfunction when compared to current treatments using standalone DOX.


Assuntos
Neoplasias da Mama/metabolismo , Doxorrubicina/farmacologia , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Oligopeptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Transdução de Sinais/efeitos dos fármacos
7.
J Nanosci Nanotechnol ; 15(9): 6413-22, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26690867

RESUMO

Triple negative breast cancer exhibit increased IL-6 expression compared with matched healthy breast tissue and a strong link between inflammation and cancer progression and metastasis has been reported. We investigated whether doxorubicin-hyaluronan-super-paramagnetic iron oxide nanoparticles (DOX-HA-SPION) would show greater therapeutic efficacy in human triple negative breast cancer cells (TNBC) MDA-MB-231, as was recently shown in drug-sensitive and multi-drug-resistant ovarian cancer cells. Therefore, we measured cellular DOX uptake via confocal microscopy; observed morphologic changes: mitochondrial and nuclear changes with electron microscopy, and quantitated apoptosis using FACS analysis after Annexin V and PI staining in MDA-MB-231 cells treated with either DOX alone or DOX-HA-SPION. We also measured both proinflammatory and anti-inflammatory cytokines; IL-6, IL-10 respectively and also measured nitrate levels in the conditioned medium by ELISA. Inaddition, NF-κB activity was measured by luciferase assay. Confocal microscopy demonstrated greater cytoplasmic uptake of DOX-HA-SPION than free DOX. We also demonstrated reduction of Vimentin with DOX-HA-SPION which is significantly less than both control and DOX. DOX-HA-SPION enhanced apoptosis and significantly down regulated both pro-inflammatory mediators IL-6 and NF-κB in comparison to DOX alone. The secretion levels of anti-inflammatory mediators IL-10 and nitrate was not decreased in the DOX or DOX-HA-SPION treatment groups. Our data suggest that DOX-HA-SPION nanomedicine-based drug delivery could have promising potential in treating metastasized and chemoresistant breast cancer by enhancing the drug efficacy and minimizing off-target effects.


Assuntos
Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Doxorrubicina/farmacocinética , Ácido Hialurônico/química , Interleucina-6/metabolismo , Nanopartículas de Magnetita/química , NF-kappa B/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Ácido Hialurônico/farmacocinética , Nanopartículas de Magnetita/toxicidade
8.
JAMA Surg ; 148(11): 1037-42, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24068167

RESUMO

IMPORTANCE: Short bowel syndrome occurs when a shortened intestine cannot absorb sufficient nutrients or fluids. Teduglutide is a recombinant analogue of human glucagonlike peptide 2 that reduces dependence on parenteral nutrition in patients with short bowel syndrome by promoting enterocytic proliferation, increasing the absorptive surface area. However, enterocyte function depends not only on the number of cells that are present but also on differentiated features that facilitate nutrient absorption and digestion. OBJECTIVE: To test the hypothesis that teduglutide impairs human intestinal epithelial differentiation. DESIGN AND SETTING: We investigated the effects of teduglutide in the modulation of proliferation and differentiation in human Caco-2 intestinal epithelial cells at a basic science laboratory. This was an in vitro study using Caco-2 cells, a human-derived intestinal epithelial cell line commonly used to model enterocytic biology. EXPOSURE: Cells were exposed to teduglutide or vehicle control. MAIN OUTCOMES AND MEASURES: We analyzed the cell cycle by bromodeoxyuridine incorporation or propidium iodide staining and flow cytometry and measured cell proliferation by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. We used quantitative reverse transcription-polymerase chain reaction to assay the expression of the enterocytic differentiation markers villin, sucrase-isomaltase, glucose transporter 2 (GLUT2), and dipeptidyl peptidase 4 (DPP-4), as well as that of the putative differentiation signals schlafen 12 (SLFN12) and caudal-related homeobox intestine-specific transcription factor (Cdx2). Villin promoter activity was measured by a luciferase-based assay. RESULTS: The MTS assay demonstrated that teduglutide increased cell numbers by a mean (SD) of 10% (2%) over untreated controls at a maximal 500 nM (n = 6, P < .05). Teduglutide increased bromodeoxyuridine-positive cells vs untreated controls by a mean (SD) of 19.4% (2.3%) vs 12.0% (0.8%) (n = 6, P < .05) and increased the S-phase fraction by flow cytometric analysis. Teduglutide reduced the mean (SD) expression of villin by 29% (6%), Cdx2 by 31% (10%), DPP-4 by 15% (6%), GLUT2 by 40% (11%), SLFN12 by 61% (14%), and sucrase-isomaltase by 28% (8%) (n = 6, P < .05 for all). CONCLUSIONS AND RELEVANCE: Teduglutide increased Caco-2 proliferation but tended to inhibit intestinal epithelial differentiation. The effects of mitogenic stimulation with teduglutide in patients with short bowel syndrome might be greater if the more numerous teduglutide-treated cells could be stimulated toward a more fully differentiated phenotype.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Peptídeos/farmacologia , Biomarcadores/metabolismo , Células CACO-2/citologia , Células CACO-2/efeitos dos fármacos , Células CACO-2/metabolismo , Técnicas de Cultura de Células , Dipeptidil Peptidase 4/metabolismo , Enterócitos/fisiologia , Transportador de Glucose Tipo 2/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Complexo Sacarase-Isomaltase/metabolismo
9.
Am J Surg ; 204(5): 602-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23140827

RESUMO

BACKGROUND: Traumatic brain injury induces a neuroinflammatory response frequently associated with increased intracranial pressure. The aim of this study was to investigate the effects of alcohol and increased extracellular pressure on murine BV-2 microglial proliferation and cytokine responses to lipopolysaccharide (LPS) stimulation. METHODS: BV-2 cells were cultured under 0 or 30 mm Hg increased extracellular pressure without or with ethanol (100 mmol/L) or LPS (10 ng/mL) for 24 hours. Cell proliferation was assessed using MTS assay and secretion of the proinflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-6, and monocyte chemotactic protein (MCP)-1 by enzyme-linked immunosorbent assay. RESULTS: Increased pressure and LPS stimulation each promoted proliferation. Ethanol pretreatment blocked these effects. Basal TNF-α and IL-6 secretion was at the limits of delectability. Basal MCP-1 production was stimulated by pressure, which was blocked by ethanol. Even this low LPS dose stimulated microglial secretion of TNF-α, IL-6, and MCP-1. Pressure inhibited LPS-stimulated production of these proinflammatory cytokines, while ethanol pretreatment blocked LPS-stimulated cytokine production. The combination of pressure and ethanol further reduced TNF-α, IL-6, and MCP-1 secretion by LPS-stimulated microglial cells. CONCLUSION: Alcohol's anti-inflammatory effects may contribute to the reduced mortality from traumatic brain injury that some have described in acutely intoxicated patients, while pressure down-regulation of inflammatory cytokine release could create a negative feedback that ameliorates inflammation in traumatic brain injury.


Assuntos
Etanol/farmacologia , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Pressão/efeitos adversos , Animais , Biomarcadores/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Ensaio de Imunoadsorção Enzimática , Interleucina-6/metabolismo , Camundongos , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Am J Physiol Cell Physiol ; 301(5): C1224-38, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21849669

RESUMO

Repetitive deformation enhances intestinal epithelial migration across tissue fibronectin. We evaluated the contribution of RhoA and its effectors Rho-associated kinase (ROK/ROCK) and mammalian diaphanous formins (mDia1) to deformation-induced intestinal epithelial motility across fibronectin and the responsible focal adhesion kinase (FAK), extracellular signal-regulated kinase (ERK), p38, and myosin light chain (MLC) signaling. We reduced RhoA, ROCK1, ROCK2, and mDia1 by smart-pool double-stranded short-interfering RNAs (siRNA) and pharmacologically inhibited RhoA, ROCK, and FAK in human Caco-2 intestinal epithelial monolayers on fibronectin-coated membranes subjected to 10% repetitive deformation at 10 cycles/min. Migration was measured by wound closure. Stimulation of migration by deformation was prevented by exoenzyme C3, Y27632, or selective RhoA, ROCK1, and ROCK2 or mDia1 siRNAs. RhoA, ROCK inhibition, or RhoA, ROCK1, ROCK2, mDia1, and FAK reduction by siRNA blocked deformation-induced nuclear ERK phosphorylation without preventing ERK phosphorylation in the cytoplasmic protein fraction. Furthermore, RhoA, ROCK inhibition or RhoA, ROCK1, ROCK2, and mDia1 reduction by siRNA also blocked strain-induced FAK-Tyr(925), p38, and MLC phosphorylation. These results suggest that RhoA, ROCK, mDia1, FAK, ERK, p38, and MLC all mediate the stimulation of intestinal epithelial migration by repetitive deformation. This pathway may be an important target for interventions to promote mechanotransduced mucosal healing during inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Sistema de Sinalização das MAP Quinases , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , ADP Ribose Transferases/farmacologia , Amidas/farmacologia , Toxinas Botulínicas/farmacologia , Células CACO-2 , Movimento Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibronectinas/metabolismo , Forminas , Humanos , Mecanotransdução Celular , Cadeias Leves de Miosina/metabolismo , Piridinas/farmacologia , RNA Interferente Pequeno/metabolismo , Quinases Associadas a rho/antagonistas & inibidores
11.
Surgery ; 150(5): 887-96, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21676422

RESUMO

BACKGROUND: Intracranial hypertension complicates severe traumatic brain injury frequently and might be associated with poor outcomes. Traumatic brain injury induces a neuroinflammatory response by microglial activation and upregulation of proinflammatory cytokines, such as interleukin-1ß, tumor necrosis factor alpha, and interleukin-6. To elucidate the effect of increased intracranial pressure on microglial function, we studied the effects of increased extracellular pressure on primary human microglial cell phagocytosis, proliferation, cytokine secretion, and total nitrate production. In addition, because many patients receive propofol during anesthesia or intensive care unit sedation, we evaluated whether propofol alters the effects of pressure. METHODS: Human microglial cells were pretreated with (2.5-20 µg/mL) propofol or Intralipid as a vehicle control were incubated at ambient atmospheric pressure or at 15 or 30 mm Hg increased pressure for 2 h for phagocytosis assays or 24 h for proliferation, cytokine secretion, and total nitrate production studies. Phagocytosis was determined by incorporation of intracellular fluorescent latex beads. Tumor necrosis factor alpha, interleukin-1ß, and interleukin-6 were assayed by sandwich enzyme-linked immunosorbent assay and total nitrate by Greiss reagent. RESULTS: Increased extracellular pressure stimulated phagocytosis versus untreated microglial cells or cells treated with an Intralipid vehicle control. Propofol also stimulated microglial phagocytosis at ambient pressure. Increased pressure, however, decreased phagocytosis in the presence of propofol. Pressure also increased microglial tumor necrosis factor-α and interleukin-1ß secretion and propofol pretreatment blocked the pressure-stimulated effect. Interleukin-6 production was not altered either by pressure or by propofol. Pressure also induced total nitrate secretion, and propofol pretreatment decreased basal as well as pressure-induced microglial nitrate production. CONCLUSION: Extracellular pressures consistent with increased intracranial pressure after a head injury activate inflammatory signals in human primary microglial cells in vitro, stimulating phagocytosis, proliferation, tumor necrosis factor-α, interleukin-1ß, and total nitrate secretion but not affecting interleukin-6. Such inflammatory events may contribute to the worsened prognosis of traumatic brain injury after increased intracranial pressure. Because propofol alleviated these potentially proinflammatory effects, these results suggest that the inflammatory cascade activated by intracranial pressure might be targeted by propofol in patients with increased intracranial pressure after traumatic brain injury.


Assuntos
Citocinas/metabolismo , Hipertensão Intracraniana/tratamento farmacológico , Microglia/efeitos dos fármacos , Nitratos/metabolismo , Fagocitose/efeitos dos fármacos , Propofol/farmacologia , Anestésicos Intravenosos/farmacologia , Bioensaio/métodos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Hipertensão Intracraniana/metabolismo , Hipertensão Intracraniana/patologia , Microglia/citologia , Microglia/metabolismo , Oxigênio/farmacologia , Pressão , Fator de Necrose Tumoral alfa/metabolismo
12.
J Surg Res ; 156(2): 217-23, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19691992

RESUMO

BACKGROUND: Repetitive deformation enhances intestinal epithelial migration across tissue fibronectin (tFN) via Src but inhibits migration across collagen. Since cell spreading generally precedes motility, we compared the effects of cyclic strain on Caco-2 spreading and migration on tFN, collagen-I, and plasma fibronectin (pFN), and investigated the role of Src in deformation-influenced spreading and migration. MATERIALS AND METHODS: Human Caco-2 intestinal epithelial cells on tFN, collagen-I or pFN were subjected to an average 10% strain at 10 cycles/min for 2 h. Src was inhibited with 10muM PP2 or Src was reduced with siRNA. Parallel studies assessed deformation effects on monolayer wound closure. RESULTS: Deformation, Src-inhibition or reduction each inhibited spreading on tFN but Src-inhibition or reduction prevented further inhibition of spreading by deformation without preventing further inhibition of motility. Deformation did not alter spreading on collagen-I or pFN, but inhibited wound closure. CONCLUSIONS: Although cell spreading generally precedes and parallels motility, repetitive deformation regulates motility independently of spreading. Since deformation activates Src, the ability of Src blockade to mimic strain-associated inhibition of spreading on tFN suggests that this effect occurs by a separate mechanism that may also require basal Src activity. Further delineation of the mechanisms by which strain disparately modulates spreading and motility may permit acceleration of mucosal healing by targeted interventions to separately promote spreading and epithelial motility.


Assuntos
Movimento Celular/efeitos dos fármacos , Matriz Extracelular/fisiologia , Mucosa Intestinal/fisiologia , Estresse Mecânico , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Células CACO-2 , Movimento Celular/fisiologia , Colágeno Tipo I/farmacologia , Matriz Extracelular/química , Fibronectinas/farmacologia , Humanos , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Quinases da Família src/antagonistas & inibidores
13.
Am J Physiol Gastrointest Liver Physiol ; 296(4): G876-85, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19179620

RESUMO

Repetitive strain stimulates intestinal epithelial migration across fibronectin via focal adhesion kinase (FAK), Src, and extracellular signal-related kinase (ERK) although how these signals act and interact remains unclear. We hypothesized that PI3K is central to this pathway. We subjected Caco-2 and intestinal epithelial cell-6 cells to 10 cycles/min deformation on flexible fibronectin-coated membranes, assayed migration by wound closure, and signaling by immunoblots. Strain stimulated PI3K, AKT, glycogen synthase kinase (GSK), and p38 phosphorylation. Blocking each kinase prevented strain stimulation of migration. Blocking PI3K prevented strain-stimulated ERK and p38 phosphorylation. Blocking AKT did not. Downstream, blocking PI3K, AKT, or ERK inhibited strain-induced GSK-Ser9 phosphorylation. Upstream of AKT, reducing FAK or Rac1 by siRNA blocked strain-stimulated AKT phosphorylation, but inhibiting Src by PP2 or siRNA did not. Transfection with FAK point mutants at Tyr397, Tyr576/577, or Tyr925 demonstrated that only FAK925 phosphorylation is required for strain-stimulated AKT phosphorylation. Myosin light chain activation by strain required FAK, Rac1, PI3K, AKT, GSK, and ERK but not Src or p38. Finally, blebbistatin, a nonmuscle myosin II inhibitor, blocked the motogenic effect of strain downstream of myosin light chain. Thus strain stimulates intestinal epithelial migration across fibronectin by a complex pathway including Src, FAK, Rac1, PI3K, AKT, GSK, ERK, p38, myosin light chain, and myosin II.


Assuntos
Movimento Celular/fisiologia , Células Epiteliais/fisiologia , Fibronectinas/fisiologia , Mucosa Intestinal/citologia , Animais , Linhagem Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Quinases da Glicogênio Sintase/antagonistas & inibidores , Quinases da Glicogênio Sintase/metabolismo , Humanos , Mecanotransdução Celular , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Biol Chem ; 284(4): 2001-11, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19047055

RESUMO

The intestinal epithelium is repetitively deformed by shear, peristalsis, and villous motility. Such repetitive deformation stimulates the proliferation of intestinal epithelial cells on collagen or laminin substrates via ERK, but the upstream mediators of this effect are poorly understood. We hypothesized that the phosphatidylinositol 3-kinase (PI3K)/AKT cascade mediates this mitogenic effect. PI3K, AKT, and glycogen synthase kinase-3beta (GSK-3beta) were phosphorylated by 10 cycles/min strain at an average 10% deformation, and pharmacologic blockade of these molecules or reduction by small interfering RNA (siRNA) prevented the mitogenic effect of strain in Caco-2 or IEC-6 intestinal epithelial cells. Strain MAPK activation required PI3K but not AKT. AKT isoform-specific siRNA transfection demonstrated that AKT2 but not AKT1 is required for GSK-3beta phosphorylation and the strain mitogenic effect. Furthermore, overexpression of AKT1 or an AKT chimera including the PH domain and hinge region of AKT2 and the catalytic domain and C-tail of AKT1 prevented strain activation of GSK-3beta, but overexpression of AKT2 or a chimera including the PH domain and hinge region of AKT1 and the catalytic domain and C-tail of AKT2 did not. These data delineate a role for PI3K, AKT2, and GSK-3beta in the mitogenic effect of strain. PI3K is required for both ERK and AKT2 activation, whereas AKT2 is sequentially required for GSK-3beta. Furthermore, AKT2 specificity requires its catalytic domain and tail region. Manipulating this pathway may prevent mucosal atrophy and maintain the mucosal barrier in conditions such as ileus, sepsis, and prolonged fasting when peristalsis and villous motility are decreased and the mucosal barrier fails.


Assuntos
Quinases da Glicogênio Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Estresse Fisiológico/fisiologia , Linhagem Celular , Proliferação de Células , Colágeno/metabolismo , Quinases da Glicogênio Sintase/antagonistas & inibidores , Humanos , Isoenzimas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miosinas/antagonistas & inibidores , Miosinas/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Mecânico , Quinases da Família src/metabolismo
15.
Am J Physiol Cell Physiol ; 294(6): C1350-61, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400991

RESUMO

Repetitive deformation due to villous motility or peristalsis may support the intestinal mucosa, stimulating intestinal epithelial proliferation under normal circumstances and restitution in injured and inflamed mucosa rich in tissue fibronectin. Cyclic strain enhances Caco-2 and IEC-6 intestinal epithelial cell migration across fibronectin via ERK. However, the upstream mediators of ERK activation are unknown. We investigated whether Src and FAK mediate strain-induced ERK phosphorylation and migration in human Caco-2 intestinal epithelial cells on fibronectin. Monolayers on tissue fibronectin-precoated membranes were subjected to an average 10% repetitive deformation at 10 cycles/min. Phosphorylation of Src-Tyr 418, FAK-Tyr 397-Tyr 576-Tyr 925, and ERK were significantly increased by deformation. The stimulation of wound closure by strain was prevented by Src blockade with PP2 (10 micromol/l) or specific short interfering (si)RNA. Src inhibition also prevented strain-induced FAK phosphorylation at Tyr 397 and Tyr 576 but not FAK-Tyr 925 or ERK phosphorylation. Reducing FAK by siRNA inhibited strain-induced ERK phosphorylation. Transfection of NH2-terminal tyrosine phosphorylation-deficient FAK mutants Y397F, Y576F-Y577F, and Y397F-Y576F-Y577F did not prevent the activation of ERK2 by cyclic strain, but a FAK mutant at the COOH terminal (Y925F) prevented the strain-induced activation of ERK2. Although the Y397F-Y576F-Y577F FAK construct exhibited less basal FAK-Tyr 925 phosphorylation under static conditions, it nevertheless exhibited increased FAK-Tyr 925 phosphorylation in response to strain. These results suggest that repetitive deformation stimulates intestinal epithelial motility across fibronectin in a manner that requires both Src activation and a novel Src-independent FAK-Tyr 925-dependent pathway that activates ERK. This pathway may be an important target for interventions to promote mucosal healing in settings of intestinal ileus or fasting.


Assuntos
Movimento Celular , Células Epiteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Mucosa Intestinal/metabolismo , Mecanotransdução Celular , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Quinases da Família src/metabolismo , Células CACO-2 , Movimento Celular/efeitos dos fármacos , Forma Celular , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Fibronectinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Adesões Focais/efeitos dos fármacos , Adesões Focais/enzimologia , Motilidade Gastrointestinal , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Mecanotransdução Celular/efeitos dos fármacos , Mutação , Fosforilação , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , Estresse Mecânico , Fatores de Tempo , Transfecção , Cicatrização , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
16.
Am J Physiol Cell Physiol ; 292(5): C1701-13, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17215324

RESUMO

Pulmonary epithelial cells are exposed to repetitive deformation during physiological breathing and mechanical ventilation. Such deformation may influence pulmonary growth, development, and barotrauma. Although deformation stimulates proliferation and activates extracellular signal-regulated kinases (ERK1/2) in human pulmonary epithelial H441 cells, the upstream mechanosensors that induce ERK activation are poorly understood. We investigated whether c-Src or focal adhesion kinase (FAK) mediates cyclic mechanical strain-induced ERK1/2 activation and proliferation in human pulmonary epithelial (NCI-H441) cells. The H441 and A549 cells were grown on collagen I-precoated membranes and were subjected to an average 10% cyclic mechanical strain at 20 cycles/min. Cyclic strain activated Src within 2 min by increasing phosphorylation at Tyr(418), followed by rapid phosphorylation of FAK at Tyr(397) and Tyr(576) and ERK1/2 at Thr(202)/Tyr(204) (n = 5, P < 0.05). Twenty-four (A549 cells) and 24-72 h (H441 cells) of cyclic mechanical strain increased cell numbers compared with static culture. Twenty-four hours of cyclic strain also increased H441 FAK, Src, and ERK phosphorylation without affecting total FAK, Src, or ERK protein. The mitogenic effect was blocked by Src (10 micromol/l PP2 or short interfering RNA targeted to Src) or MEK (50 micromol/l PD-98059) inhibition. PP2 also blocked strain-induced phosphorylation of FAK-Tyr(576) and ERK-Thr(202)/Tyr(204) but not FAK-Tyr(397). Reducing FAK by FAK-targeted short interfering RNA blocked mechanical strain-induced mitogenicity and significantly attenuated strain-induced ERK activation but not strain-induced Src phosphorylation. Together, these results suggest that repetitive mechanical deformation induced by ventilation supports pulmonary epithelial proliferation by a pathway involving Src, FAK, and then ERK signaling.


Assuntos
Proliferação de Células , Células Epiteliais/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Pulmão/metabolismo , Mecanotransdução Celular , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases da Família src/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Flavonoides/farmacologia , Quinase 1 de Adesão Focal/genética , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Mecanotransdução Celular/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Estresse Mecânico , Fatores de Tempo , Transfecção , Tirosina/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
17.
J Biol Chem ; 282(1): 14-28, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17088251

RESUMO

Intestinal epithelial cells are subject to repetitive deformation during peristalsis and villous motility, whereas the mucosa atrophies during sepsis or ileus when such stimuli are abnormal. Such repetitive deformation stimulates intestinal epithelial proliferation via focal adhesion kinase (FAK) and extracellular signal-regulated kinases (ERK). However, the upstream mediators of these effects are unknown. We investigated whether Src and Rac1 mediate deformation-induced FAK and ERK phosphorylation and proliferation in human Caco-2 and rat IEC-6 intestinal epithelial cells. Cells cultured on collagen-I were subjected to an average 10% cyclic strain at 10 cycles/min. Cyclic strain activated Rac1 and induced Rac1 translocation to cell membranes. Mechanical strain also induced rapid sustained phosphorylation of c-Src at Tyr(418), Rac1 at Ser(71), FAK at Tyr(397) and Tyr(576), and ERK1/2 at Thr(202)/Tyr(204). The mitogenic effect of cyclic strain was blocked by inhibition of Src (PP2 or short interfering RNA) or Rac1 (NSC23766). Src or Rac1 inhibition also prevented strain-induced FAK phosphorylation at Tyr(576) and ERK phosphorylation but not FAK phosphorylation at Tyr(397). Reducing FAK using short interfering RNA blocked strain-induced mitogenicity and attenuated ERK phosphorylation but not Src or Rac1 phosphorylation. Src inhibition blocked strain-induced Rac1 phosphorylation, but Rac inhibition did not alter Src phosphorylation. Transfection of a two-tyrosine phosphorylation-deficient FAK mutant Y576F/Y577F prevented activation of cotransfected myc-ERK2 by cyclic strain. Repetitive deformation induced by peristalsis or villus motility may support the gut mucosa by a pathway involving Src, Rac1, FAK, and ERK. This pathway may present important targets for interventions to prevent mucosal atrophy during prolonged ileus or fasting.


Assuntos
Células Epiteliais/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Mucosa Intestinal/metabolismo , Proteínas rac1 de Ligação ao GTP/fisiologia , Quinases da Família src/metabolismo , Animais , Células CACO-2 , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Modelos Biológicos , Mucosa/patologia , Mutação , Fosforilação , Ratos , Transdução de Sinais
18.
Kidney Int ; 68(2): 497-503, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014026

RESUMO

BACKGROUND: Oxalate is a metabolic end product excreted primarily by the kidney and associated with several pathologic conditions. The most common pathologic condition involving oxalate is the formation of calcium oxalate stones in the kidney. Several stimuli have been implicated in the development of glomerular and tubular injury in various forms of immune-mediated renal diseases. The elevated level of interleukin-6 (IL-6) has been reported in the urine of kidney stone-forming patients. In the present study, we investigated the role of oxalate, a major constituent of calcium oxalate kidney stone disease, in the production of IL-6 in normal human HK-2 kidney cells. METHODS: Confluent cultures of HK-2 cells (a renal epithelial cell line of human origin) were exposed to various concentrations of oxalate (0.2 to 2.0 mmol/L) and lipopolysaccharide (LPS) (0.1 and 10 mug/mL) for various time points (4-24 h) under serum-free conditions. The conditioned mediums were collected, and an IL-6 protein level was measured by enzyme-linked immunosorbent assay (ELISA). The total cellular RNA was isolated from the cells and subjected to relative quantitative reverse transcription-polymerase chain reaction (RT-PCR) to determine the expression of IL-6 mRNA. The statistical analysis of the results was carried out using the Student t test. RESULTS: HK-2 cells express IL-6 mRNA and protein. Oxalate increased the secretion of IL-6 protein in HK-2 cells in a concentration-dependent fashion. Oxalate exposure to HK-2 cells also induced transcriptional up-regulation of the IL-6 gene, as determined by the increased level of IL-6 mRNA expression following treatment with oxalate. Moreover, the effects of oxalate on IL-6 expression were time- and concentration-dependent. This is the first report demonstrating the regulation of IL-6 by oxalate. CONCLUSION: This study provides the first direct evidence that oxalate up-regulates the expression and secretion of IL-6 in renal epithelial cells. The increased IL-6 expression and secretion by renal epithelial cells may play a critical role in the progression of urolithiasis in hyperoxaluric conditions.


Assuntos
Hiperoxalúria/fisiopatologia , Interleucina-6/genética , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/fisiologia , Oxalatos/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Túbulos Renais Proximais/citologia , Lipopolissacarídeos/farmacologia , RNA Mensageiro/análise , Regulação para Cima/efeitos dos fármacos , Cálculos Urinários/fisiopatologia
19.
J Biol Chem ; 277(39): 36845-52, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12121971

RESUMO

Interaction of calcium oxalate monohydrate (COM) crystals with renal cells has been shown to result in altered gene expression, DNA synthesis, and cell death. In the current study the role of a stress-specific p38 MAP kinase-signaling pathway in mediating these effects of COM crystals was investigated. Exposure of cells to COM crystals (20 microg/cm(2)) rapidly stimulated strong phosphorylation and activation of p38 mitogen-activated protein kinase (p38 MAP kinase) and re-initiation of DNA synthesis. Inhibition of COM crystal binding to the cells by heparin blocked the effects of COM crystals on p38 MAPK activation. We also show that specific inhibition of p38 MAPK by 4-(4-fluorophenyl)-2-(4-methylsulfonylphenyl)-5-(4-pyridyl) imidazole (SB203580) or by overexpression of a dominant negative mutant of p38 MAP kinase abolishes COM crystal-induced re-initiation of DNA synthesis. The inhibition is dose-dependent and correlates with in situ activity of native p38 MAP kinase, determined as mitogen-activated protein kinase-activated protein kinase-2 (MAPKAP kinase-2) activity in cell extracts. In summary, inhibiting activation of p38 MAPK pathway abrogated the DNA synthesis in response to COM crystals. These data are the first demonstrations of activation of the p38 MAPK signaling pathway by COM crystals and suggest that, in response to COM crystals, this pathway transduces critical signals governing the re-initiation of DNA synthesis in renal epithelial cells.


Assuntos
Oxalato de Cálcio/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Linhagem Celular , DNA/biossíntese , DNA/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Genes Dominantes , Imidazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Suínos , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
20.
J Urol ; 168(1): 253-9, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12050552

RESUMO

PURPOSE: Oxalate, a metabolic end product, is a major constituent of majority of renal stones. Previous studies with LLC-PK1 cells, a line of proximal renal epithelial cells of porcine origin, have shown that oxalate produces time and concentration dependent effects on the growth and viability of these cells. We assessed the possibility that oxalate may be toxic to HK-2 cells, a line of human proximal renal epithelial cells. MATERIALS AND METHODS: HK-2 cells were maintained in Dulbecco's modified Eagle's medium supplemented with fetal bovine serum and antibiotics. Cells were exposed to oxalate for various intervals. Trypan blue exclusion criteria were used to assess membrane integrity, cell morphology was assessed by hematoxylin and eosin staining and crystal violet staining was used to measure cell density. DNA synthesis was measured by [3H]-thymidine incorporation and superoxide production was measured by the nitroblue tetrazolium reduction method. RESULTS: Exposure of HK-2 cells to oxalate produced time and concentration dependent increase in the membrane permeability to trypan blue and changes in the light microscopic appearance of the cells. Long-term exposure to oxalate resulted in an increase in DNA synthesis and alterations in cell viability with net cell loss after exposure to high oxalate concentrations. CONCLUSIONS: To our knowledge the results provide the first direct demonstration of the toxic effects of oxalate in HK-2 cells, a line of human renal epithelial cells, and suggest that hyperoxaluria may contribute to renal tubular damage associated with calcium oxalate stone disease.


Assuntos
Células Epiteliais/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Oxalatos/toxicidade , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Radicais Livres/metabolismo , Humanos , Túbulos Renais Proximais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...