Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 11(2)2021 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-33672590

RESUMO

Catalpol isolated from Rehmannia glutinosa is a potent antioxidant and investigated against many disorders. This review appraises the key molecular pathways of catalpol against diabetes mellitus and its complications. Multiple search engines including Google Scholar, PubMed, and Science Direct were used to retrieve publications containing the keywords "Catalpol", "Type 1 diabetes mellitus", "Type 2 diabetes mellitus", and "diabetic complications". Catalpol promotes IRS-1/PI3K/AKT/GLUT2 activity and suppresses Phosphoenolpyruvate carboxykinase (PEPCK) and Glucose 6-phosphatase (G6Pase) expression in the liver. Catalpol induces myogenesis by increasing MyoD/MyoG/MHC expression and improves mitochondria function through the AMPK/PGC-1α/PPAR-γ and TFAM signaling in skeletal muscles. Catalpol downregulates the pro-inflammatory markers and upregulates the anti-inflammatory markers in adipose tissues. Catalpol exerts antioxidant properties through increasing superoxide dismutase (sod), catalase (cat), and glutathione peroxidase (gsh-px) activity in the pancreas and liver. Catalpol has been shown to have anti-oxidative, anti-inflammatory, anti-apoptosis, and anti-fibrosis properties that in turn bring beneficial effects in diabetic complications. Its nephroprotective effect is related to the modulation of the AGE/RAGE/NF-κB and TGF-ß/smad2/3 pathways. Catalpol produces a neuroprotective effect by increasing the expression of protein Kinase-C (PKC) and Cav-1. Furthermore, catalpol exhibits a cardioprotective effect through the apelin/APJ and ROS/NF-κB/Neat1 pathway. Catalpol stimulates proliferation and differentiation of osteoblast cells in high glucose condition. Lastly, catalpol shows its potential in preventing neurodegeneration in the retina with NF-κB downregulation. Overall, catalpol exhibits numerous beneficial effects on diabetes mellitus and diabetic complications.


Assuntos
Antioxidantes/farmacologia , Diabetes Mellitus/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Glucosídeos Iridoides/farmacologia , Animais , Antioxidantes/metabolismo , Diferenciação Celular , Proliferação de Células , Complicações do Diabetes , Homeostase , Humanos , Hipoglicemiantes/farmacologia , Fígado/metabolismo , Camundongos , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/biossíntese , Doenças Neurodegenerativas/tratamento farmacológico , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Pâncreas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Rehmannia/metabolismo , Retina/efeitos dos fármacos , Transdução de Sinais
2.
Biomolecules ; 11(2)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540642

RESUMO

Golgi protein 73 (GP73) is upregulated in a variety of liver diseases, yet the detailed mechanism is poorly characterized. We analyzed GP73 in a retrospective cohort including 4211 patients with chronic liver disease (CLD) or hepatocellular carcinoma (HCC). The effect of deoxycholic acid (DCA) and nuclear factor-kappa B (NF-κB) on expression and release of GP73 in Huh-7 and SMMC7721 cells were studied. A mouse study was used to confirm our findings in vivo. A positive correlation was found between serum GP73 and total bile acid (TBA) in cirrhotic patients (r = 0.540, p < 0.001), higher than that in non-cirrhotic CLD (r = 0.318, p < 0.001) and HCC (r = 0.353, p < 0.001) patients. In Huh-7 and SMMC7721 cells, DCA upregulated the expression and release of GP73 in a dose- and time-dependent manner. After overexpressing NF-κB p65, the promoter activity, GP73 messenger RNA (mRNA) level, and supernatant GP73 level were increased. The promotion effect of DCA on GP73 release was attenuated after inhibiting the NF-κB pathway. Mutating the binding sites of NF-κB in the sequence of the GP73 promoter led to a declined promoting effect of DCA on GP73. The upregulation role of DCA in GP73 expression through the NF-κB pathway was confirmed in vivo. In addition, exposure to DCA caused disassembly of Golgi apparatus. In summary, DCA upregulates the expression and release of GP73 via activating the NF-κB pathway and destroying the Golgi structure.


Assuntos
Ácido Desoxicólico/metabolismo , Hepatopatias/metabolismo , Proteínas de Membrana/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Regulação para Cima , Adulto , Animais , Ácidos e Sais Biliares/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Doença Crônica , Ácido Desoxicólico/farmacologia , Feminino , Fibrose , Perfilação da Expressão Gênica , Humanos , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Hepatopatias/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Pessoa de Meia-Idade , Fosfoproteínas/biossíntese , Estudos Retrospectivos
3.
Cytokine ; 138: 155394, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33310423

RESUMO

Inflammation is one of the main mechanisms of pancreatic ß-cell damage and the development of type 1 diabetes (T1D). Carvedilol, a beta-adrenergic receptor blocker, has been demonstrated to have anti-inflammatory and antioxidant effects. The aim of this study was to investigate the protective effect of carvedilol against pancreatic ß-cell damage and the development of T1D in an experimental model. T1D was induced in mice by multiple low-dose streptozotocin (STZ) injections. Diabetic mice were treated with carvedilol (15 and 20 mg/kg/day, orally) for 14 days. Results showed that blood glucose levels, diabetes incidence, body weight loss and insulitis in the pancreatic tissue were significantly reduced in mice treated with carvedilol. Treatment of mice with carvedilol significantly increased the levels of antioxidants glutathione (GSH), superoxide dismutase (SOD), and catalase and decreased the levels of malondialdehyde (MDA), nitric oxide (NO) and myeloperoxidase (MPO) in the pancreatic tissue as compared with those in the STZ-induced diabetic mice. Carvedilol decreased the expression of nuclear factor kappa B (NF-κB), cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) as important modulators of inflammation and ß-cell damage in the pancreatic tissue. In addition, carvedilol significantly reduced the levels of proinflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6 IL-12, IL-17, interferon (IFN)-γ and chemokine MCP-1, while increased the anti-inflammatory cytokine IL-10 in the pancreatic tissue. In conclusion, these findings suggest that carvedilol is able to prevent pancreatic ß-cell damage and the development of T1D in mice by the inhibition of inflammatory and oxidative mediators.


Assuntos
Carvedilol/farmacologia , Ciclo-Oxigenase 2/biossíntese , Diabetes Mellitus Tipo 1/prevenção & controle , Inflamação/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Subunidade p50 de NF-kappa B/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Estresse Oxidativo , Animais , Glicemia/metabolismo , Peso Corporal , Citocinas/biossíntese , Citocinas/metabolismo , Glutationa/metabolismo , Insulina/sangue , Masculino , Malondialdeído/metabolismo , Camundongos , Pâncreas/metabolismo
4.
Histol Histopathol ; 36(1): 101-112, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33215396

RESUMO

OBJECTIVE: To examine the effect of the BRD4 inhibitor JQ1 on mice with chronic obstructive pulmonary disease (COPD) via NF-κB. METHODS: COPD models constructed by exposure to cigarette smoke and intratracheal instillation of lipopolysaccharides (LPS) in mice were treated with JQ1 (15, 25 or 50 mg/kg). HE staining was performed to observe histopathological changes in the lung tissues. Enzyme-linked immunosorbent assays (ELISAs) were used to measure the levels of IL-10, IFN-γ, IL-17, IL-1ß, IL-6, TNF-α, MMP-2, MMP-9, MDA, SOD, T-AOC and HO-1, and gelatin zymography assays were used to examine MMP-2 and MMP-9 activity. A TransAMTM NF-κB p65 detection kit was used to test NF-κB p65/DNA binding activity. Western blotting was conducted to analyze NF-κB p65 in the nucleus and its acetylation. RESULTS: JQ1 dose-dependently improved the histopathological changes in the lung tissues and decreased the mean linear intercept (MLI), destructive index and inflammatory score of the mice with COPD. The mice with COPD showed increased levels of MMP-2, MMP-9, IFN-γ, IL-17, IL-1ß, IL-6 and TNF-α with decreased IL-10 level; these changes were reversed by JQ1 in a dose-dependent manner. In addition, JQ1 reduced the MDA level and increased the SOD, HO-1 and T-AOC levels in mice with COPD, with suppression of NF-κB p65 expression in the nucleus, NF-κB/p65 (Lys310) acetylation and NF-κB p65/DNA binding activity in the lung tissues. CONCLUSION: The BRD4 inhibitor JQ1 can downregulate MMP-2 and MMP-9 expression, reduce inflammatory responses, and alleviate oxidative stress in mice with COPD, and this mechanism might be related to the inhibition of NF-κB.


Assuntos
Azepinas/farmacologia , Subunidade p50 de NF-kappa B/biossíntese , Proteínas Nucleares/antagonistas & inibidores , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/genética , Fatores de Transcrição/antagonistas & inibidores , Triazóis/farmacologia , Animais , Citocinas/metabolismo , Regulação da Expressão Gênica , Inflamação , Leucócitos/citologia , Lipopolissacarídeos/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Nucleares/genética , Estresse Oxidativo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Testes de Função Respiratória , Transdução de Sinais
5.
Int J Mol Sci ; 22(1)2020 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-33374326

RESUMO

Previous preclinical studies have demonstrated the otoprotective effects of resveratrol (RV) at low doses. This study aimed to investigate the dose-dependent effects of RV in rats with cisplatin (CXP)-induced hearing loss. Sprague-Dawley rats (8-weeks old) were divided into six treatment groups (n = 12/group) and treated as follows: control, 0.5 mg/kg RV, 50 mg/kg RV, CXP, 0.5 mg/kg RV + CXP), and 50 mg/kg RV + CXP groups. CXP (3 mg/kg) was intraperitoneally injected for 5 days. RV (0.5 or 50 mg/kg) was intraperitoneally injected for 10 days from the first day of CXP administration. Auditory brainstem response (ABR) thresholds were measured before and within 3 days at the end of the drug administration. Cochlear tissues were harvested, and the outer hair cells were examined using cochlear whole mounts. The mRNA expression of NFκB, IL6, IL1ß, and CYP1A1, and protein levels of aryl hydrocarbon receptor (AhR) and cytosolic and nuclear receptor for advanced glycation endproducts (RAGE) were evaluated. The ABR threshold increased in the 50 mg/kg RV and CXP groups at 4, 8, 16, and 32 kHz. The 0.5 mg/kg RV + CXP group demonstrated decreased hearing thresholds at 4 and 32 kHz compared to the CXP group. Cochlear whole-mount analysis revealed loss of outer hair cells in the 50 mg/kg RV and CXP groups and partial prevention of these cells in the 0.5 mg/kg RV + CXP group. The mRNA expressions of NFκB, IL6, and IL1ß were increased in the 50 mg/kg RV and CXP groups compared to the control group. In contrast, these levels were decreased in the 0.5 mg/kg RV + CXP group compared to the CXP group. The mRNA expression of CYP1A1 was increased in the CXP group, while it was decreased in the 0.5 mg/kg RV + CXP group compared to the control group. The protein levels of AhR and cytosolic RAGE decreased in the 0.5 mg/kg RV group. Low-dose RV had partial otoprotective effects on CXP ototoxicity. The otoprotective effects of RV may be mediated through anti-oxidative (CYP1A1 and RAGE) and anti-inflammatory (NFκB, IL6, and IL1ß) responses. High-dose RV exerted an inflammatory response and did not ameliorate CXP-induced ototoxicity.


Assuntos
Cisplatino/efeitos adversos , Perda Auditiva Neurossensorial/tratamento farmacológico , Resveratrol/farmacologia , Animais , Antioxidantes/farmacologia , Cóclea/efeitos dos fármacos , Citocromo P-450 CYP1A1/biossíntese , Relação Dose-Resposta a Droga , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Perda Auditiva Neurossensorial/induzido quimicamente , Inflamação , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Hidrocarboneto Arílico/metabolismo
6.
Exp Oncol ; 41(2): 90-94, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31262162

RESUMO

Asafoetida resin has been reported for various biological activities but its use has been widely restricted owing to its pungent smell and pool water solubility. AIM: In vitro study of the anticancer potential of microwave-extracted essential oil (EO) of Ferula asafoetida. MATERIALS AND METHODS: The phytochemical investigation and in vitro cytotoxicity assessment was carried out in two human liver cancer cell lines. The expression of NFKB1, TGFB1, TNF, CASP3 was analyzed by reverse transcription polymerase chain reaction. RESULTS: Ferula asafoetida EO contains high concentrations of dithiolane, which possess antiproliferative activity in human liver carcinoma cell lines (HepG2 and SK-Hep1) in a dose-dependent manner. The bioactive compounds in F. asafoetida are capable of induction of apoptosis and altered NF-kB and TGF-ß signalling with increase in caspase-3 and TNF-α expression. CONCLUSION: Further elucidation of bioactive molecules and underlying mechanisms could lead to potential intervention in liver cancer in animal models. The safety and efficacy as well as the mode of EO action in animal models would be highly crucial.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Óleos Voláteis/farmacologia , Óleos de Plantas/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Caspase 3/biossíntese , Linhagem Celular Tumoral , Ferula/química , Células Hep G2 , Humanos , NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/biossíntese , Proteínas/metabolismo , Resinas Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/metabolismo
7.
Mol Immunol ; 72: 92-102, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27011037

RESUMO

The differentiation and proper function of thymic epithelial cells (TECs) depend on various tumor necrosis factor superfamily (TNFSF) signals that are needed to maintain the thymic stromal microenvironment. Nevertheless, the direct transcriptional effects of these signals on TECs remain unclear. To address this issue, we stimulated murine embryonic thymus tissue with selected TNFSF ligands and performed a gene expression profiling study. We show that Aire expression is a direct and specific effect of RANKL stimulation, whereas LTß and TNFα are major inducers of chemokines in the thymic stroma and we propose differential NF-κB binding as one possible cause of these gene expression patterns. Our work provides further insight into the complex molecular pathways that shape the thymic microenvironment and maintain central tolerance.


Assuntos
Microambiente Celular , Células Estromais/citologia , Timo/citologia , Fatores de Necrose Tumoral/fisiologia , Animais , Perfilação da Expressão Gênica , Receptor beta de Linfotoxina/biossíntese , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/genética , Técnicas de Cultura de Órgãos , Ligante RANK/genética , Transdução de Sinais , Timo/embriologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Fatores de Necrose Tumoral/genética
8.
Turk J Haematol ; 33(1): 8-14, 2016 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25912249

RESUMO

OBJECTIVE: Nuclear factor kappa B (NF-κB) is an important transcription factor in cancer and NF-κB activation has been seen in angiogenesis, tumor progression, and metastasis. Relationships between specific NF-κB gene networks, leukemogenesis, and radiation exposure are still unknown. Our aim was to study the expression levels of the NF-κB1, NF-κB2, and Rel genes in hematological malignancies in the post-Chernobyl period. MATERIALS AND METHODS: We analyzed gene expression levels of NF-κB1, NF-κB2, and Rel in 49 B-cell chronic lymphocytic leukemia, 8 B-cell non-Hodgkin's lymphoma, 3 acute myeloid leukemia, 3 chronic myeloid leukemia, 2 hairy cell leukemia, 2 myelodysplastic syndrome, and 2 T-cell large granular lymphocytic leukemia patients using real-time polymerase chain reaction. RESULTS: Expression levels of NF-κB1, NF-κB2, and Rel genes were found to be deregulated. CONCLUSION: These results could be accepted as specific gene traces to radiation-induced leukemia or as potential candidates for new diagnostic biomarker studies. Larger experiments and non-exposed control malignant cell populations are needed to clarify these suggestions.


Assuntos
Acidente Nuclear de Chernobyl , Genes rel , Leucemia Induzida por Radiação/genética , Linfoma/genética , Subunidade p50 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/genética , NF-kappa B/genética , Neoplasias Induzidas por Radiação/genética , Fator de Transcrição RelA/genética , Adulto , Idoso , Feminino , Humanos , Leucemia Induzida por Radiação/epidemiologia , Leucemia Induzida por Radiação/etiologia , Linfoma/epidemiologia , Linfoma/etiologia , Linfoma/metabolismo , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/epidemiologia , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p52 de NF-kappa B/biossíntese , Neoplasias Induzidas por Radiação/epidemiologia , Neoplasias Induzidas por Radiação/etiologia , Neoplasias Induzidas por Radiação/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição RelA/biossíntese , Ucrânia/epidemiologia , Adulto Jovem
9.
Life Sci ; 144: 129-37, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26620764

RESUMO

AIM: Inflammatory bowel disease (IBD) is an inflammatory disorder, characterized by abnormally increased expression of Toll-like receptors TLR2 and TLR4 in the colon and increased pro-inflammatory cytokine production by macrophages. MAIN METHODS: In the present study, we explored the effect of FC-99, a novel benzenediamine analog, on dextran sulfate sodium (DSS)-induced mouse colitis and investigated its potential mechanism. KEY FINDINGS: The results revealed that FC-99 improved the colon morphology and the clinical parameters in DSS-induced mouse colitis. FC-99 inhibited the increase of DSS-induced T helper cells (Th) 1 and Th17 and enhanced the number of regulatory T cells (Treg) in mesenteric lymph nodes (MLN), but had no effect on Th2 cells. FC-99 also suppressed the DSS-induced secretion of interleukin (IL)-1ß, IL-6, and the tumor necrosis factor (TNF)-α in the colon and hindered the infiltration of macrophages into colon lamina propria. Flow cytometric analysis also confirmed that FC-99 reduced CD11b(+)F4/80(+) colon macrophages, and down-regulated TNF-α level in situ. Moreover, FC-99 inhibited concentration-dependently the expression of TNF-α and IL-6 in vitro from mouse peritoneal macrophages, which were induced by TLR ligands: PamCSK4 and peptidoglycan (PGN, TLR2 ligand) as well as LPS (TLR4 ligand). Of note, FC-99 also suppressed the activation of TLR2 and TLR4 signaling pathways and the downstream nuclear factor-κB (NF-κB) in the DSS-induced mouse colitis. SIGNIFICANCE: FC-99 improved the condition of DSS-induced mouse colitis by inhibiting the activation of TLR2 and TLR4 signaling pathways in macrophage. These results suggest that FC-99 may be developed as a new therapeutic drug for IBD.


Assuntos
Alcanossulfonatos/farmacologia , Fluorocarbonos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Colo/patologia , Sulfato de Dextrana , Feminino , Interleucina-6/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Subunidade p50 de NF-kappa B/biossíntese , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores
10.
Cancer Res ; 75(21): 4538-47, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26420215

RESUMO

Overexpression of the antiviral DNA cytosine deaminase APOBEC3B has been linked to somatic mutagenesis in many cancers. Human papillomavirus infection accounts for APOBEC3B upregulation in cervical and head/neck cancers, but the mechanisms underlying nonviral malignancies are unclear. In this study, we investigated the signal transduction pathways responsible for APOBEC3B upregulation. Activation of protein kinase C (PKC) by the diacylglycerol mimic phorbol-myristic acid resulted in specific and dose-responsive increases in APOBEC3B expression and activity, which could then be strongly suppressed by PKC or NF-κB inhibition. PKC activation caused the recruitment of RELB, but not RELA, to the APOBEC3B promoter, implicating noncanonical NF-κB signaling. Notably, PKC was required for APOBEC3B upregulation in cancer cell lines derived from multiple tumor types. By revealing how APOBEC3B is upregulated in many cancers, our findings suggest that PKC and NF-κB inhibitors may be repositioned to suppress cancer mutagenesis, dampen tumor evolution, and decrease the probability of adverse outcomes, such as drug resistance and metastasis.


Assuntos
Citidina Desaminase/biossíntese , Neoplasias/metabolismo , Proteína Quinase C/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelB/metabolismo , Linhagem Celular Tumoral , Citidina Desaminase/genética , Humanos , Antígenos de Histocompatibilidade Menor , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p52 de NF-kappa B/biossíntese , Neoplasias/genética , Infecções por Papillomavirus/patologia , Regiões Promotoras Genéticas/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Transdução de Sinais , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelB/antagonistas & inibidores , Ativação Transcricional
11.
Sci Rep ; 5: 13002, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26269414

RESUMO

Signaling by hormonal vitamin D, 1,25-dihydroxyvitamin D (1,25D) has attracted increasing interest because of its non-classical actions, particularly its putative anticancer properties and its role in controlling immune system function. Notably, the hormone-bound vitamin D receptor (VDR) suppresses signaling by pro-inflammatory NF-κB transcription factors, although the underlying mechanisms have remained elusive. Recently, the VDR was shown to enhance the turnover of the oncogenic transcription factor cMYC mediated by the E3 ligase and tumor suppressor FBW7. As FBW7 also controls the turnover of the p100 (NF-κB2) subunit of the family, we determined whether the 1,25D enhanced FBW7-dependent turnover of NF-κB subunits p100, p105 (NF-κB1) and p65 (RELA). Protein levels of all three subunits declined markedly in the presence of 1,25D in multiple cell lines in the absence of substantial changes in mRNA expression. The VDR coimmunoprecipitated with all three subunits, and 1,25D treatment accelerated subunit turnover in cycloheximide-treated cells. Importantly, we observed an association of FBW7 with p105 and p65, as well as p100, and knockdown of FBW7 eliminated 1,25D-dependent subunit turnover. Moreover, expression of NF-κB target genes was elevated in FBW7-depleted cells. These results reveal that 1,25D signaling suppresses NF-κB function by enhancing FBW7-dependent subunit turnover.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p52 de NF-kappa B/biossíntese , Receptores de Calcitriol/genética , Fator de Transcrição RelA/biossíntese , Ubiquitina-Proteína Ligases/genética , Calcitriol/genética , Calcitriol/metabolismo , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Proteína 7 com Repetições F-Box-WD , Regulação da Expressão Gênica , Células HT29 , Hormônios/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , NF-kappa B/biossíntese , NF-kappa B/genética , Subunidade p50 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Receptores de Calcitriol/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Ubiquitina-Proteína Ligases/metabolismo , Vitamina D/administração & dosagem , Vitamina D/análogos & derivados , Vitamina D/genética , Vitamina D/metabolismo
12.
J Biol Chem ; 290(30): 18391-9, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26037922

RESUMO

The Werner syndrome helicase (WRN) plays a role in maintaining genomic stability. The lack of WRN results in Werner syndrome, a rare autosomal recessive genetic disorder, which causes premature aging accompanied by many complications such as rare forms of cancer and type 2 diabetes. However, the underlying mechanisms of these complications, arising due to the loss of WRN, are poorly understood. In this study, we demonstrated the function of WRN in transcriptional regulation of NF-κB targets. WRN physically interacts via its RecQ C-terminal (RQC) domain with the Rel homology domain of both the RelA (p65) and the p50 subunits of NF-κB. In the steady state, WRN is recruited to HIV-1 long terminal repeat (LTR), a typical NF-κB-responsive promoter, as well as the p50/p50 homodimer, in an NF-κB site-dependent manner. The amount of WRN on LTR increased along with the transactivating RelA/p50 heterodimer in response to TNF-α stimulation. Further, a knockdown of WRN reduced the transactivation of LTR in exogenous RelA/p50-introduced or TNF-α-stimulated cells. Additionally, knockdown of WRN reduced TNF-α stimulation-induced activation of the endogenous promoter of IL-8, an NF-κB-responsive gene, and WRN increased its association with the IL-8 promoter region together with RelA/p50 after TNF-α stimulation. In conjunction with studies that have shown NF-κB to be a key regulator of aging and inflammation, our results indicate a novel role of WRN in transcriptional regulation. Along with NF-κB, the loss of WRN is expected to result in incorrect regulation of downstream targets and leads to immune abnormalities and homeostatic disruption.


Assuntos
Exodesoxirribonucleases/genética , Interleucina-8/genética , Subunidade p50 de NF-kappa B/biossíntese , RecQ Helicases/genética , Fator de Transcrição RelA/biossíntese , Síndrome de Werner/genética , Envelhecimento/genética , Envelhecimento/patologia , Exodesoxirribonucleases/metabolismo , HIV-1/genética , Células HeLa , Humanos , Interleucina-8/biossíntese , Subunidade p50 de NF-kappa B/genética , Regiões Promotoras Genéticas , RecQ Helicases/metabolismo , Fator de Transcrição RelA/genética , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia , Síndrome de Werner/patologia , Helicase da Síndrome de Werner
13.
Tumour Biol ; 36(8): 6029-36, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25736926

RESUMO

Epidermal growth factor receptor (EGFR) and its downstream elements are overexpressed in most cases of the head and neck squamous cell carcinoma. This study investigated the expression pattern of key proteins linked to the EGFR pathway in laryngeal carcinoma patients with a history of cannabis smoking. We selected 83 male glottic cancer patients, aged between 45 to 75 years with three distinct populations-nonsmoker, cigarette smoker, and cannabis smoker. Immunohistochemical staining was performed for EGFR, protein kinase B (PKB or Akt), nuclear factor kappa B p50 (NF-КB), and cyclooxygenase-2 (COX-2) followed by boolean scoring for statistical analysis. Experimental data showed upregulation of the selected EGFR cascade in tumor cells, stromal expression of EGFR, and nuclear localization of COX-2 in metaplastic gland cells of laryngeal cancer tissue sample. Statistical analyses indicated that overexpression of the EGFR cascade is significantly correlated to cannabis smoking. Cannabis smokers had higher expression (p < 0.01) of these onco-proteins with respect to both nonsmokers as well as cigarette smokers. Risk factor analysis showed high risk of these proteins expression in age <60 years (odds ratio (OR) > 1.5) as the lower age group had relatively higher number of cannabis smokers. This study provides evidence for a direct association between cannabis smoking and increased risk of laryngeal cancer. Higher expression of the EGFR cascade in cannabis smokers revealed that cannabis smoking may be a major cause for the early onset of aggressive laryngeal cancer.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Receptores ErbB/biossíntese , Neoplasias Laríngeas/genética , Subunidade p50 de NF-kappa B/biossíntese , Proteína Oncogênica v-akt/biossíntese , Idoso , Ciclo-Oxigenase 2/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Laríngeas/induzido quimicamente , Neoplasias Laríngeas/patologia , Masculino , Fumar Maconha/efeitos adversos , Pessoa de Meia-Idade , Subunidade p50 de NF-kappa B/genética , Proteína Oncogênica v-akt/genética , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos
14.
Cell Prolif ; 48(2): 221-30, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25736627

RESUMO

OBJECTIVES: This study aimed to investigate effects of neurotrophin receptor-mediated melanoma antigen-encoding gene homology (NRAGE) on proliferation and odontoblastic differentiation of mouse dental pulp cells (mDPCs). MATERIALS AND METHODS: Mouse dental pulp cells were infected with recombinant lentivirus to stably knockdown expression of NRAGE, and biological effects of NRAGE on the cells were detected. Proliferation and odontoblastic differentiation of mDPCs were observed. Simultaneously, mRNA and protein levels of NRAGE and nuclear factor-κB (NF-κB) protein expression were detected. Immunofluorescence assay was used to detect expression and location of NRAGE and NF-κB. RESULTS: NRAGE mRNA and protein levels reduced significantly after mDPC odontoblastic induction. Knockdown of NRAGE inhibited the proliferation of mDPCs. However, knockdown of NRAGE enhanced their odontoblastic differentiation with up-regulated ALPase activity. It also promoted mineral nodule formation as well as mRNA and protein expressions of ALP, DSPP and DMP1. Protein levels of NF-κB/p50 significantly increased, whereas NF-κB/p105 protein expression decreased in the mDPC/shNRG group. Immunofluorescence revealed that relocation of NF-κB was similar to that of NRAGE during odontoblastic induction, in which NF-κB translocated from the cytoplasm to the nucleus. CONCLUSION: NRAGE is a potent regulator of proliferation and odontoblastic differentiation of mDPCs, which might be via the NF-κB signalling pathway.


Assuntos
Diferenciação Celular/genética , Polpa Dentária/citologia , Proteínas de Neoplasias/genética , Odontoblastos/citologia , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Proliferação de Células/genética , Células Cultivadas , Proteínas da Matriz Extracelular/biossíntese , Proteínas de Membrana/biossíntese , Camundongos , Subunidade p50 de NF-kappa B/biossíntese , Proteínas de Neoplasias/biossíntese , Fosfoproteínas/biossíntese , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno , Sialoglicoproteínas/biossíntese
15.
Free Radic Biol Med ; 82: 1-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25614461

RESUMO

Evidence suggests that upregulation of soluble epoxide hydrolase (sEH) is associated with the development of myocardial infarction, dilated cardiomyopathy, cardiac hypertrophy, and heart failure. However, the upregulation mechanism is still unknown. In this study, we treated H9C2 cells with buthionine sulfoximine (BSO) to explore whether oxidative stress upregulates sEH gene expression and to identify the molecular and cellular mechanisms behind this upregulatory response. Real-time PCR and Western blot analyses were used to measure mRNA and protein expression, respectively. We demonstrated that BSO significantly upregulated sEH at mRNA levels in a concentration- and time-dependent manner, leading to a significant increase in the cellular hypertrophic markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Furthermore, BSO significantly increased the cytosolic phosphorylated IκB-α and translocation of NF-κB p50 subunits, as measured by Western blot analysis. This level of translocation was paralleled by an increase in the DNA-binding activity of NF-κB P50 subunits. Moreover, our results demonstrated that pretreatment with the NF-κB inhibitor PDTC significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression in a dose-dependent manner. Additionally, mitogen-activated protein kinases (MAPKs) were transiently phosphorylated by BSO treatment. To understand further the role of MAPKs pathway in BSO-mediated induction of sEH mRNA, we examined the role of extracellular signal-regulated kinase (ERK), c-JunN-terminal kinase (JNK), and p38 MAPK. Indeed, treatment with the MEK/ERK signal transduction inhibitor, PD98059, partially blocked the activation of IκB-α and translocation of NF-κB p50 subunits induced by BSO. Moreover, pretreatment with MEK/ERK signal transduction inhibitors, PD98059 and U0126, significantly inhibited BSO-mediated induction of sEH and cellular hypertrophic marker gene expression. These results clearly demonstrated that the NF-κB signaling pathway is involved in BSO-mediated induction of sEH gene expression, and appears to be associated with the activation of the MAPK pathway. Furthermore, our findings provide a strong link between sEH-induced cardiac dysfunction and involvement of NF-κB in the development of cellular hypertrophy.


Assuntos
Butionina Sulfoximina/farmacologia , Cardiomegalia/patologia , Epóxido Hidrolases/biossíntese , Sistema de Sinalização das MAP Quinases/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Animais , Antioxidantes/farmacologia , Fator Natriurético Atrial/biossíntese , Butadienos/farmacologia , Linhagem Celular , Sobrevivência Celular , Ativação Enzimática , Epóxido Hidrolases/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Regulação da Expressão Gênica/fisiologia , Glutationa/biossíntese , Insuficiência Cardíaca/patologia , Proteínas I-kappa B/biossíntese , Proteínas I-kappa B/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mioblastos Cardíacos/metabolismo , Inibidor de NF-kappaB alfa , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Subunidade p50 de NF-kappa B/biossíntese , Peptídeo Natriurético Encefálico/biossíntese , Nitrilas/farmacologia , Estresse Oxidativo , Fosforilação , Prolina/análogos & derivados , Prolina/farmacologia , RNA Mensageiro/biossíntese , Ratos , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
PLoS One ; 9(10): e111087, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333282

RESUMO

BACKGROUND: Recruitment of bone marrow derived endothelial progenitor cells (BMDEPCs) alleviates multiple organ injury (MOI) and improves outcomes. However, mechanisms mediating BMDEPC recruitment following septic MOI remain largely unknown. This study characterized the kinetics of BMDEPC recruitment and proliferation and defined the role of NF-κB in regulating BMDEPC recruitment and proliferation. METHODS AND MAIN FINDINGS: Chimeric mice with an intact or disrupted NF-κB p50 gene and BMDEPC-restricted expression of green fluorescent protein were created and injected with LPS (2 mg/kg, i.p.). BMDEPC recruitment and proliferation in multiple organs were quantified. BMDEPC recruitment and proliferation are highly organ-dependent. Lungs had the highest number of BMDEPC recruitment, whereas heart, liver and kidney had only a small fraction of the number of BMDEPCs in lungs. Number of proliferating BMDEPCs was several-fold higher in lungs than in other 3 organs. Kinetically, BMDEPC recruitment into different organs showed different time course profiles. NF-κB plays obligatory roles in mediating BMDEPC recruitment and proliferation. Universal deletion of NF-κB p50 gene inhibited LPS-induced BMDEPC recruitment and proliferation by 95% and 69% in heart. However, the contribution of NF-κB to these regulations varies significantly between organs. In liver, universal p50 gene deletion reduced LPS-induced BMDEPC recruitment and proliferation only by 49% and 35%. NF-κB activities in different tissue compartments play distinct roles. Selective p50 gene deletion either in stromal/parenchymal cells or in BM/blood cells inhibited BMDEPC recruitment by a similar extent. However, selective p50 gene deletion in BM/blood cells inhibited, but in stromal/parenchymal cells augmented BMDEPC proliferation. CONCLUSIONS: BMDEPC recruitment and proliferation display different kinetics in different organs following endotoxemic MOI. NF-κB plays obligatory and organ-dependent roles in regulating BMDEPC recruitment and proliferation. NF-κB activities in different tissue compartments play distinct roles in regulating BMDEPC proliferation.


Assuntos
Endotoxemia/genética , Insuficiência de Múltiplos Órgãos/genética , Traumatismo Múltiplo/genética , Subunidade p50 de NF-kappa B/biossíntese , NF-kappa B/genética , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Endotoxemia/induzido quimicamente , Endotoxemia/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Camundongos , Insuficiência de Múltiplos Órgãos/induzido quimicamente , Insuficiência de Múltiplos Órgãos/patologia , Traumatismo Múltiplo/induzido quimicamente , Traumatismo Múltiplo/patologia , Subunidade p50 de NF-kappa B/genética
17.
PLoS One ; 9(9): e108278, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25265479

RESUMO

BACKGROUND: The core protein of hepatitis C virus (HCV) is found in the cytoplasm and nuclei of infected cells, including hepatocytes and other cells in the liver. The core protein could be secreted as well. Resident liver macrophages are dependent on the tissue micro-environment and external stimuli to differentiate M1 and M2 hypotypes with distinct functions, and increased expression of the nuclear transcription factor STAT3 was seen in M2-polarized macrophages. In contrast to proinflammatory M1 macrophages, M2 macrophages serve beneficial roles in chronic inflammation, immunosuppression, and tumorigenesis. METHODS: Monocyte-derived human macrophage line (mTHP-1) was treated with the exogenous HCV core protein. Next, the mTHP-1 culture supernatant or cell pellets were added to culture media of normal human liver cell line (L02). RESULTS: Only the culture supernatant stimulated L02 cells proliferation, which was associated with phosphorylated ERK expression. Core protein activated mTHP-1 cells showed enhanced pro- and anti-inflammatory cytokines secretion, which was accompanied by high expression of phosphorylated NF-κB105 and NF-κB65. However, phosphorylated STAT1, and STAT3, which are normally associated with M1 and M2 macrophage polarization, and cell surface expression of CD206, CD14, CD16, and CD86, were unaltered. A transwell co-culture system showed that only in mTHP-1 co-cultured with L02 in the presence of exogenous core protein, were higher levels of phosphorylated STAT3 and CD206 seen. CONCLUSIONS: We showed L02 cells proliferation was accelerated by the culture supernatant of mTHP-1 cells treated with the exogenous HCV core protein. The exogenous core protein mediated the interaction between macrophages and hepatocytes in co-culture, which enhanced the expression of phosphorylated STAT3 and CD206 in macrophages.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Hepatócitos/fisiologia , Macrófagos/fisiologia , Proteínas Recombinantes/farmacologia , Proteínas do Core Viral/farmacologia , Antígeno B7-2/biossíntese , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Proteínas Ligadas por GPI/biossíntese , Hepacivirus/genética , Humanos , Lectinas Tipo C/biossíntese , Receptores de Lipopolissacarídeos/biossíntese , Receptor de Manose , Lectinas de Ligação a Manose/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Fosforilação , Receptores de Superfície Celular/biossíntese , Receptores de IgG/biossíntese , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/biossíntese , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição RelA/biossíntese
18.
Cytokine ; 69(1): 14-21, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25022957

RESUMO

Tumor necrosis factor (TNF)-α is a major effector in various inflammatory conditions. TNF-like weak inducer of apoptosis (TWEAK) is a member of the TNF superfamily that promotes inflammatory tissue damage through its receptor, FGF-inducible molecule 14 (Fn14). Since both TWEAK and TNF-α have been shown to mediate pathological responses through inter-dependent or independent pathways by in vitro, the potential interplay of these pathways was investigated in a mouse colitis model. Acute colitis was induced by rectal injection of trinitrobenzene sulfonic acid (TNBS), with administration of control IgG, TNF receptor (TNFR)-Ig chimeric protein, anti-TWEAK monoclonal antibody, or the combination of TNFR-Ig and anti-TWEAK antibody. On day 4, disease severity was evaluated and gene expression profiling was analyzed using whole colon tissue. NF-κB activation was investigated with Western blot. Levels of transcript of TWEAK, Fn14 and NF-κB-related molecules were measured in purified colon epithelial cells (ECs). As a result, activation of the canonical (p50/RelA), but not noncanonical (p100/RelB)-mediated pathway was the hallmark of inflammatory responses in this model. Inflammation induced upregulation of Fn14 only in ECs but not in other cell types. Combination treatment of TNFR-Ig and anti-TWEAK antibody synergistically reduced disease severity in comparison with the control antibody or single agent treatment. Gene expression profile of the colon indicated downregulation of canonical NF-κB pathway with combination treatment. In conclusion, synergistic activation of canonical NF-κB by TWEAK and TNF-α is critical for the induction of inflammatory tissue damage in acute inflammation.


Assuntos
Colite/patologia , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Colite/induzido quimicamente , Colo/citologia , Colo/patologia , Citocina TWEAK , Perfilação da Expressão Gênica , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/citologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Receptores do Fator de Necrose Tumoral/biossíntese , Receptor de TWEAK , Fator de Transcrição RelA/biossíntese , Ácido Trinitrobenzenossulfônico
19.
Mol Cell Biol ; 34(16): 2961-80, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24891615

RESUMO

Epithelial to mesenchymal transition (EMT) is activated during cancer invasion and metastasis, enriches for cancer stem cells (CSCs), and contributes to therapeutic resistance and disease recurrence. Signal transduction kinases play a pivotal role as chromatin-anchored proteins in eukaryotes. Here we report for the first time that protein kinase C-theta (PKC-θ) promotes EMT by acting as a critical chromatin-anchored switch for inducible genes via transforming growth factor ß (TGF-ß) and the key inflammatory regulatory protein NF-κB. Chromatinized PKC-θ exists as an active transcription complex and is required to establish a permissive chromatin state at signature EMT genes. Genome-wide analysis identifies a unique cohort of inducible PKC-θ-sensitive genes that are directly tethered to PKC-θ in the mesenchymal state. Collectively, we show that cross talk between signaling kinases and chromatin is critical for eliciting inducible transcriptional programs that drive mesenchymal differentiation and CSC formation, providing novel mechanisms to target using epigenetic therapy in breast cancer.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/genética , Isoenzimas/genética , Proteína Quinase C/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Antígeno CD24/biossíntese , Antígeno CD24/genética , Diferenciação Celular/genética , Cromatina/genética , Montagem e Desmontagem da Cromatina/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Células MCF-7 , Subunidade p50 de NF-kappa B/biossíntese , Subunidade p50 de NF-kappa B/genética , Invasividade Neoplásica , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Proteína Quinase C-theta , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transdução de Sinais/genética , Esferoides Celulares/patologia , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/genética , Fator de Crescimento Transformador beta/genética
20.
Biomed Res Int ; 2014: 178410, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24963474

RESUMO

NF-κB signaling promotes cancer progression in a large number of malignancies. Metadherin, a coactivator of the NF-κB transcription complex, was recently identified to regulate different signaling pathways that are closely related to cancer. We assessed the immunohistochemical expression of p50, p65, and metadherin in 30 ovarian carcinomas, 15 borderline ovarian tumours, and 31 benign ovarian cystadenomas. Ovarian carcinomas exhibited significantly higher expression of all 3 markers compared to benign ovarian tumours. Borderline ovarian tumours demonstrated significantly higher expression for all 3 markers compared to benign cystadenomas. Ovarian carcinomas demonstrated significantly higher expression of p50 and metadherin compared to borderline ovarian tumours, whereas no significant difference was noted in p65 expression between ovarian carcinomas and borderline ovarian tumours. There was a strong correlation with the expression levels of p50, p65, and metadherin, whereas no correlation was observed with either grade or stage. Strong p50, p65, and metadherin expression was associated with a high probability to distinguish ovarian carcinomas over borderline and benign ovarian tumours, as well as borderline ovarian tumours over benign ovarian neoplasms. A gradual increase in the expression of these molecules is noted when moving across the spectrum of ovarian carcinogenesis, from borderline ovarian tumours to epithelial carcinomas.


Assuntos
Moléculas de Adesão Celular/biossíntese , Regulação Neoplásica da Expressão Gênica , Subunidade p50 de NF-kappa B/biossíntese , Proteínas de Neoplasias/biossíntese , Neoplasias Ovarianas/metabolismo , Fator de Transcrição RelA/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imuno-Histoquímica/métodos , Proteínas de Membrana , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Proteínas de Ligação a RNA , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...