Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Multidiscip Healthc ; 16: 411-418, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36820220

RESUMO

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a highly contagious viral infection. In addition to its association with common pulmonary and gastrointestinal complications, COVID-19 is also associated with numerous neurological and neuropsychiatric conditions. This minireview aims to cover current literature addressing the application of telemedicine in neurological disorders and neuropsychiatric conditions, especially in response to the COVID-19 pandemic. This article revealed that quarantine, masking, and social distancing policies practiced during the COVID-19 pandemic involved restrictions and challenges to providing medical services, especially for patients with neurological disorders with or without COVID-19 infection. During the pandemic, both healthcare administrators and clinicians, including neurologists, have rapidly adapted or introduced telemedicine technologies for delivering specialty care. In some areas in the world, telemedicine has been successfully applied to reduce the impact imposed by COVID-19. Conclusively, this article supports the idea that telemedicine is an effective tool for providing specialized healthcare for patients with neurological conditions while adhering to social distancing or lockdown policies instituted during the COVID-19 pandemic. Government and medical/healthcare authorities, physicians and healthcare providers need to work together to expand the adoption of telemedicine applications, even after the COVID-19 crisis.

3.
Cancers (Basel) ; 12(5)2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32422901

RESUMO

Guanine nucleotide-binding protein-like-3-like (GNL3L) is a crucial regulator of NF-κB signaling that is aberrantly activated during diverse chemoresistance-associated cellular processes. However, the molecular mechanisms of GNL3L tumor initiation and resistant state are largely unknown. Moreover, the identification of predictive biomarkers is necessary to effectively generate therapeutic strategies for metastatic human colorectal cancer (CRC). This study aims to identify how cells acquire resistance to anticancer drugs and whether the downregulation of miR-4454 is associated with the progression of CRC. Here, we have shown that the overexpression of miR-4454 in resistant tumors is a crucial precursor for the posttranscriptional repression of GNL3L in human chemoresistant CRC progression, and we used doxycycline induced miR-4454 overexpression that significantly reduced tumor volume in a subcutaneous injection nude mice model. Together, these observations highlight that the downregulation of miR-4454 in resistant clones is prominently responsible for maintaining their resistance against anticancer drug therapy. Our study indicates that the development of miR-4454 as a microRNA-based therapeutic approach to silence GNL3L may remarkably reduce oncogenic cell survival that depends on GNL3L/NF-κB signaling, making miR-4454 a candidate for treating metastatic human CRC.

4.
Respir Physiol Neurobiol ; 271: 103284, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31472287

RESUMO

Acute respiratory distress syndrome (ARDS) is a disease with high morbidity and mortality rates. The recruitment maneuver (RM) is one of the interventions used for ARDS patients suffering from severe hypoxemia. RM works by opening the atelectatic lungs using high transpulmonary pressure. RM has therefore been widely used for many years in patients with ARDS. However, because of the high transpulmonary pressure used in this intervention, there are concerns about both biotrauma and hemodynamic instability. To assess the effects of RM in ARDS, we conducted a study using three groups of pigs (n = 6 in each group): group I (control), group II (ARDS), and group III (ARDS with RM). After measuring the baseline values, ARDS was induced by deactivating the surfactant with 5% Tweens lavage. For pigs of group III, the RM protocol used was positive end-expiratory pressure (PEEP) of 25 cmH2O and peak pressure of 45 cmH2O. Gas exchange, hemodynamics, the expression of cytokines in serum, bronchoalveolar lavage fluid (BALF), and exhaled breath condensates (EBCs) were measured. The baseline measurements taken were similar across the three groups, and no significant difference was noted. After the induction of ARDS, PaO2 substantially decreased, while PaCO2, alveolar-arterial O2 gradient, pulmonary arterial pressure, lung water, level of cytokines in serum, EBCs, and BALF all increased. After RM, gas exchange and lung water level improved, but the level of cytokines in EBCs and BALF increased. Although RM led to an improvement in gas exchange, it may cause release of inflammatory cytokines in the EBCs and BALF.


Assuntos
Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Troca Gasosa Pulmonar/fisiologia , Síndrome do Desconforto Respiratório/metabolismo , Animais , Líquido da Lavagem Broncoalveolar , Citocinas/genética , Expressão Gênica , Síndrome do Desconforto Respiratório/genética , Mecânica Respiratória/fisiologia , Suínos
5.
Cancers (Basel) ; 11(10)2019 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-31623173

RESUMO

Colorectal cancer (CRC) is the second leading cause of cancer-related illness worldwide and one of the most common malignancies. Therefore, colorectal cancer research and cases have gained increasing attention. Oxaliplatin (OXA) is currently used in first-line chemotherapy to treat stage III and stage IV metastatic CRC. However, patients undergoing chemotherapy often develop resistance to chemo drugs being used. Evidence has confirmed that microRNAs regulate downstream genes in cancer biology and thereby have roles related to tumor growth, proliferation, invasion, angiogenesis, and multi-drug resistance. The aim of our study is to establish whether miR-31-5p is an oncogene in human colorectal cancers that are resistant to OXA and further confirm its malignant phenotype-associated target molecule. From the results of miRNA microarray assay, we establish that miR-31-5p expression was upregulated in oxaliplatin-resistant (OR)-LoVo cells compared with parental LoVo cells. Moreover, through in vitro and in vivo experiments, we demonstrate that miR-31-5p and large tumor suppressor kinase 2 (LATS2) were inversely related and that miR-31-5p and Forkhead box C1 (FOXC1) were positively correlated in the same LoVo or OR-LoVo cells. Importantly, we reveal a novel drug-resistance mechanism in which the transcription factor FOXC1 binds to the miR-31 promoter to increase the expression of miR31-5p and regulate LATS2 expression, resulting in cancer cell resistance to OXA. These results suggest that miR-31-5p may be a novel biomarker involved in drug resistance progression in CRC patients. Moreover, the FOXC1/miR31-5p/LATS2 drug-resistance mechanism provides new treatment strategies for CRC in clinical trials.

6.
Cancers (Basel) ; 11(7)2019 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-31261976

RESUMO

Hepatocellular carcinoma (HCC) is a common fatal type of malignant tumor that has highly metastatic and recurrent properties. Fisetin is a natural flavonoid found in various vegetables and fruits which exhibits anti-cancer and anti-inflammatory properties, as well as other effects. Thus, we hypothesized that fisetin can act as an adjuvant therapy in cancer or drug-resistant cancer cells, and further investigated the molecular mechanisms underlying the development of drug-resistance in HCC cells. We found that fisetin effectively inhibited the cell viability of not only parental cells but also histone deacetylase inhibitors-resistant (HDACis-R) cells and enhanced the chemosensitivity of HCC cells. Interestingly, fisetin did not induce cell apoptosis through the activation of the endoplasmic reticulum (ER) stress sensor of protein kinase R (PKR)-like endoplasmic reticulum kinase, but rather through the non-canonical pathway of the protein phosphatase 1 (PP1)-mediated suppression of eIF2α phosphorylation. Moreover, fisetin-induced cell apoptosis was reversed by treatment with PP1 activator or eIF2α siRNA in HCC cells. Based on these observations, we suggest that PP1-eIF2α pathways are significantly involved in the effect of fisetin on HCC apoptosis. Thus, fisetin may act as a novel anticancer drug and new chemotherapy adjuvant which can improve the efficacy of chemotherapeutic agents and diminish their side-effects.

7.
J Cell Physiol ; 234(7): 11822-11834, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30552676

RESUMO

Cancer stem cells (CSCs) exist in colon cancer and exhibit characteristics of stem cells which are due to lineages of tissues where they arise. Epithelial to mesenchymal transition (EMT)-undergoing cancer cells display CSC properties and therapeutic resistance. Cancer and stromal cells comprise of a tumor microenvironment. One way the two populations communicate with each other is to secret CXC ligands (CXCLs). CXCLs are capable of causing chemotaxis of specific types of stromal cells and control angiogenesis. Double immunofluorescence, western blot analysis, and colony-formation assay were carried out to compare parental and CPT-11-resistant LoVo cells. CPT-11-R LoVo colon cancer cells showed increased expression of CXCL1, CXCL2, CXCL3, and CXCL8. They displayed significantly increased intracellular protein levels of CXCL2 and CXCR2. CPT-11-R LoVo cells showed significantly elevated expression in aldehyde dehydrogenase 1 (ALDH1), cluster of differentiation 24 (CD24), cluster of differentiation 44 (CD44), and epithelial cell adhesion molecule (EpCAM). CXCL2 knockdown by short hairpin RNA resulted in reduced expression of CSC proteins, cyclins, EMT markers, G proteins, and matrix metalloproteinases (MMPs). Finally, Gαi-2 was found to promote expression of CSC genes and tumorigenesis which were more apparent in the resistant cells. In addition, Gαq/11 showed a similar pattern with exceptions of EpCAM and MMP9. Therefore, CXCL2-CXCR2 axis mediates through Gαi-2 and Gαq/11 to promote tumorigenesis and contributes to CSC properties of CPT-11-R LoVo cells.


Assuntos
Quimiocina CXCL2/metabolismo , Neoplasias do Colo/patologia , Resistencia a Medicamentos Antineoplásicos , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Irinotecano/farmacologia , Células-Tronco Neoplásicas/patologia , Receptores de Interleucina-8B/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Neoplásicas/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
8.
Chin J Physiol ; 61(5): 266-279, 2018 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-30384400

RESUMO

Regulated upon activation, normal T cell expressed, and secreted (RANTES), also known as chemokine ligand 5 (CCL5), has been reported to facilitate macrophage migration, which plays a crucial role in tissue inflammation. The aim of this study is to investigate the characteristics and underlying mechanism of RANTES on macrophage chemotaxis under physiological and pathological conditions. The study was conducted on macrophage RAW264.7 cell and bone marrow-derived macrophages (BMDM) isolated from CCL receptor 5 (CCR5) knockout mice. The macrophage migration and glucose uptake was assessed in time and dose dependent manners. Moreover, reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis were used to characterize mRNA and protein level related to the underlying mechanism. The present result showed that the maraviroc, a selective CCR5 inhibitor, dose-dependently suppressed RANTES-induced rapid increases in glucose uptake and cell migration in RAW264.7 cells. Similar effects were observed in the BMDM isolated from CCR5 knockout mice compared with wild type control. RANTES treatment promptly enhanced membrane glucose transporter 1 (GLUT1) expression, glucose uptake as well as phosphorylation of AKT on Thr308, Ser473 within min and has prolonged effect on phosphorylation of AMP-activated protein kinase (AMPK) on Thr172, which were abrogated by maraviroc, CCR5 siRNA or phospholipase C (PLC) inhibitor in RAW264.7 cells. Inhibition of PI3K and AMPK by LY294002 and Compound C significantly suppress RANTES-stimulated macrophage glucose uptake and migration, respectively. RANTES has biphasic effect on activating PLC signaling including prompt action on PI3K/AKT phosphorylation and prolong action on AMPK phosphorylation via CCR5 which leads to increased GLUT1-mediated glucose uptake and macrophage migration under physiopathological states.


Assuntos
Quimiocina CCL5 , Macrófagos , Animais , Quimiotaxia , Camundongos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Receptores CCR5 , Transdução de Sinais , Linfócitos T , Fosfolipases Tipo C
9.
Cell Biochem Funct ; 36(4): 176-182, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29654619

RESUMO

ZAK is a novel mixed lineage kinase-like protein that contains a leucine-zipper and a sterile-alpha motif as a protein-protein interaction domain, and it is located in the cytoplasm. There are 2 alternatively spliced forms of ZAK: ZAKα and ZAKß. Previous studies showed that ZAKα is involved in various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy, but the molecular mechanism of ZAKß is not yet known. In a recent study in our laboratory, we found that ZAKß can ameliorate the apoptotic effect induced by ZAKα in H9c2 cells. We further hypothesized that ZAKß could also improve the apoptotic effect induced by ZAKα in human osteosarcoma cells. The results of this study show that ZAKß can induce apoptosis and decrease cell viability similar to the effects of ZAKα. Interestingly, our ZAKα-specific inhibitor assay shows that the expression of ZAKß is highly dependent on ZAKα expression. However, ZAKß expression effectively induces ZAKα expression and results in synergistic enhancement of apoptosis in human osteosarcoma cells. Furthermore, co-immunoprecipitation results revealed that ZAKα can directly interact with ZAKß, and this interaction may contribute to the enhanced apoptotic effects. SIGNIFICANCE OF THE STUDY: ZAK is a mixed lineage kinase involved in cell differentiation, proliferation, and hypertrophic growth. ZAKα isoform of ZAK is associated with tumorigenesis, but the function of ZAKß is not yet known. In H9c2 cells, ZAKß was found to ameliorate the apoptotic effect induced by ZAKα. However, in osteosarcoma cells, ZAKß elevates the apoptotic effect induced by ZAKα. In this study, we show that similar to ZAKα, the ZAKß induces apoptosis and decreases cell viability. Interestingly, the expression of ZAKß is dependent on ZAKα expression, and ZAKß further enhances ZAKα expression and results in synergistic enhancement of apoptosis in osteosarcoma cells.


Assuntos
Apoptose/efeitos dos fármacos , Osteossarcoma/metabolismo , Proteínas Quinases/biossíntese , Antibióticos Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Humanos , MAP Quinase Quinase Quinases , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
10.
J Cell Physiol ; 233(9): 7134-7142, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29574877

RESUMO

Irinotecan (CPT11) and Oxaliplatin have been used in combination with fluorouracil and leucovorin for treating colorectal cancer. However, the efficacy of these drugs is reduced due to various side effects and drug resistance. Fisetin, a hydroxyflavone possess anti-proliferative, anti-cancer, anti-inflammatory, and antioxidant activity against various types of cancers. Apart from that, fisetin has been shown to induce cytotoxic effects when combined with other known chemotherapeutic drugs. In this study, we aimed to investigate whether Fisetin was capable of sensitizing both Irinotecan and Oxaliplatin resistance colon cancer cells and explored the possible signaling pathways involved using In vitro and In vivo models. The results showed that Fisetin treatment effectively inhibited cell viability and apoptosis of CPT11-LoVo cells than Oxaliplatin (OR) and parental LoVo cancer cells. Western blot assays suggested that apoptosis was induced by fisetin administration, promoting Caspase-8, and Cytochrome-C expressions possibly by inhibiting aberrant activation of IGF1R and AKT proteins. Furthermore, fisetin inhibited tumor growth in athymic nude mouse xenograft model. Overall, our results provided a basis for Fisetin as a promising agent to treat parental as well as chemoresistance colon cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Flavonoides/farmacologia , Irinotecano/farmacologia , Oxaliplatina/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo , Flavonóis , Masculino , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Environ Toxicol ; 33(2): 191-197, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29105997

RESUMO

Human osteosarcoma (OS) is a malignant cancer of the bone. It exhibits a characteristic malignant osteoblastic transformation and produces a diseased osteoid. A previous study demonstrated that doxorubicin (DOX) chemotherapy decreases human OS cell proliferation and might enhance the relative RNA expression of ZAK. However, the impact of ZAKα overexpression on the OS cell proliferation that is inhibited by DOX and the molecular mechanism underlying this effect are not yet known. ZAK is a protein kinase of the MAPKKK family and functions to promote apoptosis. In our study, we found that ZAKα overexpression induced an apoptotic effect in human OS cells. Treatment of human OS cells with DOX enhanced ZAKα expression and decreased cancer cell viability while increasing apoptosis of human OS cells. In the meantime, suppression of ZAKα expression using shRNA and inhibitor D1771 both suppressed the DOX therapeutic effect. These findings reveal a novel molecular mechanism underlying the DOX effect on human OS cells. Taken together, our findings demonstrate that ZAKα enhances the apoptotic effect and decreases cell viability in DOX-treated human OS cells.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Doxorrubicina/toxicidade , Proteínas Quinases/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , MAP Quinase Quinase Quinases , NF-kappa B/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Proteínas Quinases/química , Proteínas Quinases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína bcl-X/metabolismo
13.
J Cell Physiol ; 233(7): 5458-5467, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29247488

RESUMO

Oxaliplatin (OXA), is a third generation platinum drug used as first-line chemotherapy in colorectal cancer (CRC). Cancer cells acquires resistance to anti-cancer drug and develops resistance. ATP-binding cassette (ABC) drug transporter ABCG2, one of multidrug resistance (MDR) protein which can effectively discharge a wide spectrum of chemotherapeutic agents out of cancer cells and subsequently reduce the intracellular concentration of these drugs. Role of ABCG2 and plausible molecular signaling pathways involved in Oxaliplatin-Resistant (OXA-R) colon cancer cells was evaluated in the present study. OXA resistant LoVo cells was developed by exposing the colon cells to OXA in a dose-dependent manner. Development of multi drug resistance in OXA-R cells was confirmed by exposing the resistance cells to oxaliplatin, 5-FU, and doxorubicin. OXA treatment resulted in G2 phase arrest in parental LoVo cells, which was overcome by OXA-R LoVo cells. mRNA and protein expression of ABCG2 and phosphorylation of NF-κB was significantly higher in OXA-R than parental cells. Levels of ER stress markers were downregulated in OXA-R than parental cells. OXA-R LoVo cells exposed to NF-κB inhibitor QNZ effectively reduced the ABCG2 and p-NF-κB expression and increased ER stress marker expression. On other hand, invasion and migratory effect of OXA-R cells were found to be decreased, when compared to parental cells. Metastasis marker proteins also downregulated in OXA-R cells. ABCG2 inhibitor verapamil, downregulate ABCG2, induce ER stress markers and induces apoptosis. In vivo studies in nude mice also confirms the same.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Neoplasias Colorretais/tratamento farmacológico , Proteínas de Neoplasias/genética , Oxaliplatina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Proliferação de Células/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Resistencia a Medicamentos Antineoplásicos/genética , Estresse do Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Oxaliplatina/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
World J Gastroenterol ; 23(7): 1171-1179, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28275297

RESUMO

AIM: To identify potential anti-cancer constituents in natural extracts that inhibit cancer cell growth and migration. METHODS: Our experiments used high dose thymoquinone (TQ) as an inhibitor to arrest LoVo (a human colon adenocarcinoma cell line) cancer cell growth, which was detected by cell proliferation assay and immunoblotting assay. Low dose TQ did not significantly reduce LoVo cancer cell growth. Cyclooxygenase 2 (COX-2) is an enzyme that is involved in the conversion of arachidonic acid into prostaglandin E2 (PGE2) in humans. PGE2 can promote COX-2 protein expression and tumor cell proliferation and was used as a control. RESULTS: Our results showed that 20 µmol/L TQ significantly reduced human LoVo colon cancer cell proliferation. TQ treatment reduced the levels of p-PI3K, p-Akt, p-GSK3ß, and ß-catenin and thereby inhibited the downstream COX-2 expression. Results also showed that the reduction in COX-2 expression resulted in a reduction in PGE2 levels and the suppression of EP2 and EP4 activation. Further analysis showed that TG treatment inhibited the nuclear translocation of ß-catenin in LoVo cancer cells. The levels of the cofactors LEF-1 and TCF-4 were also decreased in the nucleus following TQ treatment in a dose-dependent manner. Treatment with low dose TQ inhibited the COX-2 expression at the transcriptional level and the regulation of COX-2 expression efficiently reduced LoVo cell migration. The results were further verified in vivo by confirming the effects of TQ and/or PGE2 using tumor xenografts in nude mice. CONCLUSION: TQ inhibits LoVo cancer cell growth and migration, and this result highlights the therapeutic advantage of using TQ in combination therapy against colorectal cancer.


Assuntos
Benzoquinonas/farmacologia , Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Neoplasias do Colo/patologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Compostos Fitoquímicos/uso terapêutico , Transdução de Sinais
15.
Environ Toxicol ; 32(1): 62-69, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26537528

RESUMO

Arecoline, the most abundant alkaloid in betel nut is known to promote abnormal proliferation of epithelial cells by enhancing epidermal growth factor receptor (EGFR) activation and cyclooxygenase-2 (COX2) expression. Taiwanin C, a naturally occurring lignan extracted from Taiwania cryptomerioides, has been found to be a potential inhibitor of COX2 expression. Based on the MTT assay results, taiwanin C was found to be effective in inhibiting the tumorous T28 cell than the non-tumorous N28 cells. The modulations in the expression of relevant proteins were determined to understand the mechanism induced by taiwanin C to inhibit T28 cell proliferation. The levels of activated EGFR and COX2 were found to be abnormally high in the T28 oral cancer cells. However, taiwanin C was found to inhibit the activation of EGFR and regulated other related downstream proteins and thereby inhibited the T28 cell proliferation. In conclusion the results indicate that taiwanin C suppresses COX2-EGFR and enhances P27 pathways to suppress arecoline induced oral cancer cell proliferation via ERK1/2 inactivation. © 2015 Wiley Periodicals, Inc. Environ Toxicol 32: 62-69, 2017.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Antineoplásicos Fitogênicos/farmacologia , Arecolina/antagonistas & inibidores , Arecolina/toxicidade , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Lactonas/farmacologia , Lignanas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Bucais/patologia , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/biossíntese , Receptores ErbB/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
Environ Toxicol ; 32(2): 519-529, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26990689

RESUMO

In our previous experiments, we found ß-catenin was highly expressed in the tumor area with high invasive ability and poor prognosis. In this study, we have examined the mechanism by which ERα regulates ß-catenin expression as well as the metastasis ability of hepatocellular cancer HA22T cells. To identify whether the anticancer effect of estrogen and ERα is mediated through suppression of ß-catenin expression, we co-transfected pCMV-ß-catenin and ERα into HA22T cells, and determined the cell motility by wound healing, invasion, and migration assays. Results showed that estrogen and/or ERα inhibited ß-catenin gene expression and repressed HA22T cell motility demonstrated that similar data was observed in cells expressing the ERα stable clone. Moreover, we examined the protein-protein interaction between ERα and ß-catenin by immunostain, co-immunoprecipitation, and Western blotting. E2 enhanced the binding of ERα with ß-catenin and then triggered ß-catenin to bind with E3 ligase (ßTrCP) to promote ß-catenin degradation. Finally by employing systematic ChIP studies, we showed ERα can interact directly with the ß-catenin promoter region following E2 treatment. All our results reveal that estrogen and ERα blocked metastatic function of HA22T cells by modulating GSK3ß and ßTrCP expression and further enhanced ß-catenin degradation and suppressed its downstream target genes. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 519-529, 2017.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , beta Catenina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Microscopia de Fluorescência , Proteínas Contendo Repetições de beta-Transducina/genética
17.
Environ Toxicol ; 32(2): 669-678, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27060453

RESUMO

Clinically used chemotherapeutics can effectively eliminate most tumor cells. However, they cause unwanted side effects and result in chemoresistance. To overcome such problems, phytochemicals are now used to treat cancers by means of targeted therapy. Thymoquinone (TQ) is used to treat different cancers (including colon cancer) and is an NF-κB inhibitor. Irinotecan resistant (CPT-11-R) LoVo colon cancer cell line was previous constructed by step-wise CPT-11 challenges to un-treated parental LoVo cells and expresses EGFR/IKKα/ß/NF-κB pathway. TQ resulted in reduced total and phosphorylation of IKKα/ß and NF-κB and decreased metastasis in CPT-11-R cells. TQ not only reduced activity of ERK1/2 and PI3K but also activated JNK and p38. Furthermore, TQ was also found to suppress metastasis through activation of JNK and p38. Therefore, TQ suppressed metastasis through NF-κB inhibition and activation of JNK and p38 in CPT-11-R LoVo colon cancer cells. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 669-678, 2017.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Benzoquinonas/farmacologia , Camptotecina/análogos & derivados , Fator de Transcrição RelA/antagonistas & inibidores , Camptotecina/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Humanos , Irinotecano , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Pathol Oncol Res ; 23(3): 477-485, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27757837

RESUMO

Peroxisome proliferator-activated receptor-α (PPARα) is a member of the nuclear receptor superfamily involved in hepatocarcinogenesis in rodents. In previous studies on liver tumor tissues, PPARα mRNA expression was found to be significantly higher and overexpression of ERα inhibited the PPARα expression, cell-proliferation and also induced apoptosis in Hep3B cell. However, the role of ERß is not known yet. Therefore, the aim of this study is to define the role of ERß on PPARα in Hep3B cells. The effect of PPARα signaling cascade were monitored by inducing Hep3B cells by fenofibrate. Further the cells were transfected with pCMV-ERß and the consequences of ERß-overexpression on the PPARα induced changes such as enhanced cell-proliferation and suppressed apoptosis were determined using western blot analysis and TUNEL assay. The EMSA was used to identify whether ERß modulates PPARα expression by binding to PPARα promoter region to repress PPARα promoter activity. In addition, the direct interaction between ERß and PPARα proteins was verified by co-immunoprecipitation assay. Our results show that the overexpressed ERß not only attenuated the effects of fenofibrate to induce the levels of apoptosis protein such as Cyt.c, Caspase 9 and Caspase 3 but also inhibited the levels of survival protein such Bcl-xL, p-Bad, cyclin A and cyclin E. All these effects of E2/ERß resulted in the enhancement of mitochondria dependent apoptotic pathway and the attenuation of cell proliferation. Moreover, the overexpressed ERß reduced the mRNA and protein levels of PPARα and its downstream Acyl-CoA oxidase (ACO). EMSA results show that ERß directly binds to PPRE and inhibit PPARα gene expression and according to immunoprecipitation assay ERß also binds strongly with PPARα. The E2/ERß further inhibited the fenofibrate-induced nuclear translocation of PPARα. Taken together, ERß might directly downregulate PPARα gene expression and inhibit the nuclear translocation to suppress the proliferation and induce the apoptosis of Hep3B cells.


Assuntos
Antígeno 12E7/genética , Apoptose/genética , Carcinoma Hepatocelular/genética , Proliferação de Células/genética , Receptor beta de Estrogênio/genética , Neoplasias Hepáticas/genética , PPAR alfa/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Núcleo Celular/genética , Regulação para Baixo/genética , Expressão Gênica/genética , Humanos , Neoplasias Hepáticas/patologia , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Translocação Genética/genética
19.
Cell Biochem Biophys ; 74(2): 221-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27193608

RESUMO

Cytoskeleton filaments play an important role in cellular functions such as maintaining cell shape, cell motility, intracellular transport, and cell division. Actin-binding proteins (ABPs) have numerous functions including regulation of actin filament nucleation, elongation, severing, capping, cross linking, and actin monomer sequestration. Gelsolin (GSN) is one of the actin-binding proteins. Gelsolin (GSN) is one of the actin-binding proteins that regulate cell morphology, differentiation, movement, and apoptosis. GSN also regulates cell morphology, differentiation, movement, and apoptosis. In this study, we have used H9c2 cardiomyoblast cell and H9c2-GSN stable clones to understand the roles and mechanisms of GSN overexpression in hypoxia-induced cardiomyoblast cell death. The data show that hypoxia or GSN overexpression induces HIF-1α expression and reduces the expression of survival markers p-Akt and Bcl-2 in H9c2 cardiomyoblast cells. Under hypoxic conditions, GSN overexpression further reduces p-Akt expression and elevates total as well as cleaved GSN levels and HIF-1α levels. In addition, GSN overexpression enhances apoptosis in cardiomyoblasts under hypoxia. Hypoxic challenge further induced activated caspase-3 and cell death that was attenuated after GSN knock down, which implies that GSN is a critical therapeutic target against hypoxia-induced cardiomyoblast cell death.


Assuntos
Apoptose , Gelsolina/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mioblastos/citologia , Miócitos Cardíacos/citologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Biomarcadores/metabolismo , Caspase 3/metabolismo , Hipóxia Celular , Linhagem Celular , Gelsolina/deficiência , Gelsolina/genética , Técnicas de Silenciamento de Genes , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos
20.
Environ Toxicol ; 31(12): 2045-2056, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26464283

RESUMO

Helioxanthin, an active compound from Taiwania cryptomerioides Hayata, has been shown to have various biological activities. However, their anticancer effect in oral squamous cell carcinoma has not been well established yet. Helioxanthin inhibited the proliferation of oral squamous cell carcinoma cells in a dose-dependent manner by inducing G2/M phase arrest. Similarly, helioxanthin inhibited cyclooxygenase-2, (COX-2), phosphorylated EGFR, and extracellular-signal-regulated kinases (ERK) protein level and further reduced the nuclear accumulation of phosphorylated epidermal growth factor receptor (pEGFR) and activator protein-1(AP-1) family protein, c-fos. Moreover, helioxanthin at the dose of 20 and 30 mg kg-1 for 15 days reduced the tumor growth in animal model. This study demonstrated that Helioxanthin exerts its anticancer activity against oral cancer cells by downregulating EGFR/ERK/c-fos signaling pathway to inhibit COX-2 level and by activating cyclin-dependent kinase inhibitor (p27) to further induce G2/M cell cycle arrest. This helioxanthin may serve as a novel candidate for oral cancer prevention. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 2045-2056, 2016.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lignanas/farmacologia , Neoplasias Bucais/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Arecolina , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Xenoenxertos , Lignanas/uso terapêutico , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Nus , Neoplasias Bucais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA