Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Signal ; 111: 110856, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37598918

RESUMO

Vascular endothelial growth factor receptor-3 (VEGFR-3) is known to participate in tumorigenesis and lymphangiogenesis, and as such, has the potential to serve as a molecular target for cancer therapy. SAR131675 is a highly selective VEGFR-3 antagonist that has an inhibitive effect on lymphatic cell growth. However, the anticancer effects and underlying mechanisms of SAR131675 in ovarian cancer remain poorly understood. In this study, we investigated the pathological role of VEGFR-3, and the effects of SAR131675 on proliferation, cell cycle, migration, and apoptosis in ovarian cancer cells. Our results showed that the mRNA and protein of VEGFR-3 were expressed in OVCAR3 and SKOV3 ovarian cancer cells, and this receptor was activated following stimulation with 50 ng/ml VEGF-C Cys156Ser (VEGF-CS), a selective ligand for VEGFR-3. Enhancing VEGFR-3 phosphorylation by treatment of ovarian cancer cells with VEGF-CS resulted in increased levels of phosphorylated extracellular signal-regulated kinases 1/2 (ERK1/2) and AKT. Moreover, our data demonstrated that SAR131675 inhibited VEGF-CS-mediated proliferation, colony formation, and migration of cancer cells in a dose-dependent manner. In addition, inhibition of VEGFR-3 activation with SAR131675 significantly increased cell cycle arrest and promoted apoptosis in both OVCAR3 and SKOV3 cells. Mechanistically, SAR131675 effectively suppressed the VEGF-CS-induced phosphorylation of VEGFR-3 and its downstream effectors including activated ERK1/2 and AKT in ovarian cancer cells. Our results reveal an anticancer activity of SAR131675 on the growth and migration of ovarian cancer cells, which may be through inhibiting VEGFR-3/ERK1/2/AKT pathway. SAR131675 may serve as an effective targeted drug for ovarian cancer.

2.
J Cell Biochem ; 124(5): 674-686, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36922713

RESUMO

Vascular endothelial growth factor receptor 3 (VEGFR3) is expressed in cancer cell lines and exerts a critical role in cancer progression. However, the signaling pathways of VEGFR3 in ovarian cancer cell proliferation remain unclear. This study aimed to demonstrate the signaling pathways of VEGFR3 through the upregulated expression of miR-1236 in ovarian cancer cells. We found that the messenger RNA and protein of VEGFR3 were expressed in the ovarian cancer cell lines, but downregulated after microRNA-1236 (miR-1236) transfection. The inhibition of VEGFR3, using miR-1236, significantly reduced cell proliferation, clonogenic survival, migration, and invasion ability in SKOV3 and OVCAR3 cells (p < 0.01). The flow cytometry results indicated that the rate of apoptotic cells in SKOV3 (38.65%) and OVCAR3 (41.95%) cells increased following VEGFR3 inhibition. Moreover, VEGFR3 stimulation (using a specific ligand, VEGF-CS) significantly increased extracellular signal-regulated kinase 1/2 (ERK1/2) and protein kinase B (AKT) phosphorylation (p < 0.01), whereas VEGFR3 suppression reduced p-ERK1/2 (67.94% in SKOV3 and 93.52% in OVCAR3) and p-AKT (59.56% in SKOV3 and 78.73% in OVCAR3) compared to the VEGF-CS treated group. This finding demonstrated that miR-1236 may act as an endogenous regulator of ERK1/2 and AKT signaling by blocking the upstream regulator of VEGFR3. Overall, we demonstrated the important role of the miR-1236/VEGFR3 axis in ovarian cancer cell proliferation by regulating the ERK1/2 and AKT signaling that might be an effective strategy against ovarian cancer.


Assuntos
MicroRNAs , Neoplasias Ovarianas , Receptor 3 de Fatores de Crescimento do Endotélio Vascular , Feminino , Humanos , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/farmacologia
3.
Res Pharm Sci ; 17(5): 527-539, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36386487

RESUMO

Background and purpose: Quantum dots (QDs) are semiconductor nanocrystals that are widely used in biology due to their good optical properties. QDs, especially cadmium-based QDs, play an important role in the diagnosis and treatment of cancer due to their intrinsic fluorescence. The aim of the present study was the evaluation of the cellular uptake mechanisms of CdTe QDs in ovarian cancer cell lines. Experimental approach: In this study, we used CdTe QDs coated with thioglycolic acid. The ovarian cancer cell lines SKOV3 and OVCAR3 were treated with different concentrations of QDs, triamterene, chlorpromazine, and nystatin, and cell viability was evaluated through the MTT test. To find the way of cellular uptake of CdTe QDs, we used the MTT test and interfering compounds in endocytic pathways. Intrinsic fluorescence and cellular internalization of CdTe QDs were assessed using flow cytometry and fluorescence microscopy imaging. Findings / Results: The viability of CdTe QDs-treated cells dose-dependently decreased in comparison to untreated cells. To evaluate the cellular uptake pathways of CdTe QDs, in most cases, a significant difference was observed when the cells were pretreated with nystatin. The results of flow cytometry showed the cellular uptake of CdTe QDs was dose- and time-dependent. Conclusion and implications: Nystatin had a measurable effect on the cellular uptake of CdTe QDs. This finding suggests that caveola-mediated endocytosis has a large portion on the internalization of CdTe QDs. According to the results of this study, CdTe QDs may have potential applications in cancer research and diagnosis.

4.
Anticancer Agents Med Chem ; 22(5): 943-950, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34238175

RESUMO

BACKGROUND: Tyrosine Kinase Inhibitors (TKIs) can be used to inhibit cancer cell proliferation by targeting the vascular endothelial growth factor receptor (VEGFR) family. SAR131675 is a highly selective receptor tyrosine kinase inhibitor to VEGFR3 that reveals the inhibitory effect on proliferation in human lymphatic endothelial cells. However, the molecular mechanisms underlying this process are generally unclear. OBJECTIVE: This study was performed to investigate the possible involvement of the Bcl-2/Bax/Cyto c apoptosis pathway in Human Umbilical Vein Endothelial Cells (HUVECs). In addition, the role of Reactive Oxygen Species (ROS) and mitochondrial membrane potential was evaluated. METHODS: The effect of SAR131675 on HUVEC cell viability was evaluated by MTT assay. The activity of SAR131675 in inducing apoptosis was carried out through the detection of Annexin V-FITC/PI signal by flow cytometry. To determine the mechanisms underlying SAR131675 induced apoptosis, the mitochondrial membrane potential, ROS generation, the activity of caspase-3, and expression of apoptosis-related proteins such as Bcl-2, Bax, and cytochrome c were evaluated in HUVECs. RESULTS: SAR131675 significantly inhibited cell viability and induced apoptosis in HUVECs in a dose-dependent manner. Moreover, SAR131675 induced mitochondrial dysfunction, ROS generation, Bcl-2 down-regulation, Bax upregulation, cytochrome c release, and caspase-3 activation, which displays features of mitochondria-dependent apoptosis signaling pathway. CONCLUSION: Our present data demonstrated that SAR131675-induced cytotoxicity in HUVECs associated with the mitochondria apoptotic pathway. These results suggest that further studies are required to fully elucidate the role of TKIs in these cellular processes.


Assuntos
Citocromos c , Fator A de Crescimento do Endotélio Vascular , Apoptose , Caspase 3/metabolismo , Citocromos c/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias , Naftiridinas , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína X Associada a bcl-2/metabolismo
5.
Toxicol In Vitro ; 56: 184-193, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30682495

RESUMO

Cadmium (Cd) as a human carcinogen and one of the most toxic industrial and environmental pollutant mimics the estrogenic effects in cell proliferation. So, it might have a role in the incidence and etiology of hormone-related cancers such as ovarian cancer as the most lethal gynecologic malignancy. This study aimed to evaluate the estrogenic effect and underlying mechanism of Cd in ovarian cancer cell line proliferation. OVCAR3 and SKOV3 cell lines were treated with different concentrations of CdCl2 (0- 50 µM). Cell proliferation was analyzed using MTT and BrdU assay. To evaluate the estrogenic effect of Cd, the cells were pre-incubated with estrogen receptor (ER) antagonist ICI 182,780. The expression of ER was determined using western blotting method. Real-time RT-PCR method was used to assess c-fos, c-jun and FOXO3a mRNA level. The results showed that Cd has an estrogenic proliferative effect at nM concentration range and ICI 182,780 significantly reversed the CdCl2-induced cell proliferation. Cd also increased the expression of ERs. Cd exposure induced activation of p-ERK1/2 in these cells. Cd also intensified c-jun, c-fos, and FOXO3a mRNA expression. Taken together, the current work suggests that Cd induces ovarian cancer cell proliferation in an ER-dependent mechanism induced ERK1/2 activation pathway. Understanding of downstream targets by which Cd deregulates cell proliferation can be noteworthy to define its underlying carcinogenesis mechanism.


Assuntos
Cloreto de Cádmio/toxicidade , Carcinógenos/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Estrogênio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Proteína Forkhead Box O3/genética , Humanos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética
6.
Res Pharm Sci ; 13(2): 159-167, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29606970

RESUMO

Cadmium (Cd), a ubiquitous environmental and occupational pollutant, acts as a metalloestrogen to induce cell proliferation. It is suggested that Cd may also contribute to the development of estrogen-related cancers like ovarian cancer which is the most lethal cancer in women. Furthermore, it was shown that melatonin has antiproliferative effect on estradiol (E2)-induced proliferation. The aim of the present study was to evaluate whether melatonin inhibits Cd-induced proliferation in ovarian cancer cell lines and also whether Cd and melatonin can modulate estrogen receptor α (ERα) expression. OVCAR3 and SKOV3 human ovarian cancer cell lines were treated with CdCl2 (1-100 nM) and melatonin (1 µM) for 48 h. Cell proliferation evaluation was carried out by bromodeoxyuridine (BrdU) incorporation assay. ERα expression was detected by western blotting method 24 h after cell treatment. The results were demonstrated that Cd increased proliferation of ovarian cancer cell lines in a dose dependent manner. Melatonin inhibited Cd-induced proliferation of OVCAR3 and SKOV3 cell lines. Moreover, CdCl2 significantly increased ERα expression in both OVCAR3 and SKOV3 cell lines compared to control. Melatonin significantly inhibited Cd inducing effect on ERα expression of OVCAR3 and SKOV3 cell. In conclusion, due to the proliferative effect on ovarian cancer cell lines, Cd could play an important role in the etiology of ovarian cancer by inducing cells ERα expression. Furthermore, melatonin has the protective role on Cd-induced cell proliferation by inhibition of ERα expression.

7.
Toxicol Ind Health ; 34(5): 339-352, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29591890

RESUMO

INTRODUCTION: Semiconductor quantum dots (QDs), especially those containing cadmium, have undergone marked improvements and are now widely used nanomaterials in applicable biological fields. However, great concerns exist regarding their toxicity in biomedical applications. Because of the lack of sufficient data regarding the toxicity mechanism of QDs, this study aimed to evaluate the cytotoxicity of three types of QDs: CdTe QDs, high yield CdTe QDs, and CdTe/CdS core/shell QDs on two human breast cancer cell lines MDA-MB468 and MCF-7. METHODS: The breast cancer cells were treated with different concentrations of QDs, and cell viability was evaluated via MTT assay. Hoechst staining was applied for observation of morphological changes due to apoptosis. Apoptotic DNA fragmentation was visualized by the agarose gel electrophoresis assay. Flow cytometric annexin V/propidium iodide (PI) measurement was used for apoptosis detection. RESULTS: A significant decrease in cell viability was observed after QDs treatment ( p < 0.05). Apoptotic bodies and chromatin condensation was observed by Hoechst staining. DNA fragmentation assay demonstrated a DNA ladder profile in the exposed cells and also annexin V/PI flow cytometry confirmed apoptosis in a dose-dependent manner. CONCLUSION: Our results revealed that CdTe, high yield CdTe, and CdTe/CdS core/shell QDs induce apoptosis in breast cancer cell lines in a dose-dependent manner. This study would help realizing the underlying cytotoxicity mechanism, at least partly, of CdTe QDs and may provide information for the development of nanotoxicology and safe use of biological applications of QDs.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Cádmio/toxicidade , Pontos Quânticos/toxicidade , Telúrio/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Humanos , Células MCF-7
8.
Res Pharm Sci ; 13(1): 57-64, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29387112

RESUMO

Estrogen is a steroid hormone that plays a key role in the development and regulation of reproductive system. It has been shown that estrogen is related to breast cancer development through binding to its receptors. In order to uncover the estrogen effects on adenosine receptor expression, estrogen-positive MCF-7 cells were used to treat with agonist and antagonist of estrogen and then the mRNA expression of adenosine receptor subtypes were evaluated. Estrogen-positive MCF-7 cells were treated with various concentrations of 17ß estradiol (E2) as an estrogen agonist, and ICI 182,780 as an estrogen antagonist. The gene expression of adenosine receptor subtypes were detected by real time RT-PCR. The results of MTT assay showed that E2 increased cell viability in a dose dependent manner. The expression pattern of all adenosine receptor subtypes are as follow; A2b > A1 > A2a > A3 in untreated MCF-7 cells. Obtained results showed that E2 incubation at 0.001-0.01 µM led to up-regulation of A1ARs, A2aARs and A3ARs dose dependently. E2 at 0.001 µM also had no significant effect on A2bARs expression but, at higher doses induced a considerable decrease in mRNA A2bARs expression. Treatment with antagonist confirmed that up-regulation of these receptors is mediated by estrogen receptor. Taken together, our results indicate that treatment of MCF-7 cells with E2 led to up-regulation of adenosine receptors. However, these effects were partially restored by treatment with antagonist suggesting that such effects are mediated by estrogen receptors.

9.
J Cell Biochem ; 119(2): 2333-2344, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28885729

RESUMO

Leptin, an adipokine secreted by adipose tissue, induces cell invasion and metastasis. MMP7 is a member of the matrix metalloproteinase family that plays an important role in cell invasion. Here we evaluate the possible role and underlying mechanism of MMP7 in the leptin-mediated cell invasion in ovarian cancer cell lines. All experiments were carried out in cultured SKOV3, OVCAR3, and CaoV-3 ovarian cell lines. MMP7 expression was determined using the Western blot following treatment to various concentrations of leptin for defined time intervals. The activation of ERK, JNK, and P38 MAP kinases were determined using Western blotting. Wound healing and BD matrigel invasion assays were used to measure cell migration and invasion. The siRNA approach and pharmacological inhibitors of ERK and JNK pathway were used to confirm the receptor-dependent effect of leptin and a role for ERK and JNK pathway. Zymography assay was employed to determine MMP2 and MMP9 activation. Results show that leptin induces ERK1/2 and JNK1/2 activation and subsequently promotes MMP7 expression in SKOV3 (4.8 ± 0.14 fold of control, P < 0.01) and OVCAR3 (3.1 ± 0.19 fold of control, P < 0.01) ovarian cancer cell lines. These effects was reversed by knockdown of OB-Rb and/or pre-incubation with PD98059 (ERK1/2 inhibitor), SP600125 (JNK1/2 inhibitor). Gelatin zymography showed that MMP7 gene silencing attenuated leptin-induced MMP9 activation in SKOV3 cell line. Taken together, our results suggest new evidences for a modulatory effect of leptin in regulation of ovarian cancer cell invasion by stimulating MMP7 expression via ERK and JNK pathways.


Assuntos
Leptina/genética , Leptina/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Neoplasias Ovarianas/metabolismo , Antracenos/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Flavonoides/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica
10.
Cell Oncol (Dordr) ; 41(1): 61-72, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29218545

RESUMO

PURPOSE: It has been reported that cancer stem cells (CSCs) may play a crucial role in the development, recurrence and metastasis of breast cancer. Targeting signaling pathways in CSCs is considered to be a promising strategy for the treatment of cancer. Here, we investigated the role of the A2B adenosine receptor (A2BAR) and its associated signaling pathways in governing the proliferation and viability of breast cancer cell line derived CSCs. METHODS: CSCs were isolated from the breast cancer cell lines MCF-7 and MDA-MB-231 using a mammosphere assay. The effect of the A2BAR agonist BAY606583 on cell proliferation was evaluated using XTT and mammosphere formation assays, respectively. Apoptosis was assessed using Annexin-V staining and cell cycle analyses were performed using flow cytometry. The expression levels of Bax, Bcl-2, cyclin-D1, CDK-4 and (phosphorylated) ERK1/2 were assessed using Western blotting. RESULTS: Our data revealed that the breast cancer cell line derived mammospheres were enriched for CSCs. We also found that A2BAR stimulation with its agonist BAY606583 inhibited mammosphere formation and CSC viability. In addition, we found that the application of BAY606583 led to CSC cell cycle arrest and apoptosis through the cyclin-D1/Cdk-4 and Bax/Bcl-2 pathways, respectively. Notably, we found that BAY606583 significantly down-regulated ERK1/2 phosphorylation in the breast cancer cell line derived CSCs. CONCLUSIONS: From our results we conclude that A2BAR induces breast CSC cell cycle arrest and apoptosis through downregulation of the ERK1/2 cascade. As such, A2BAR may be considered as a novel target for the treatment of breast cancer.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor A2B de Adenosina/metabolismo , Aminopiridinas/farmacologia , Apoptose , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/enzimologia , Fosforilação
11.
Res Pharm Sci ; 13(6): 493-499, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30607147

RESUMO

Cadmium (Cd) as a metalloesterogen may have a role in development of ovarian cancer. One of the critical target genes of estrogens is progesterone receptors (PRs). There are controversial studies on association between Cd, PRs, and cell proliferation. This study investigates the effect of Cd on proliferation of ovarian cancer cell lines, PRA and PRB expression and their relationship. OVCAR3 and SKOV3 cells were treated with CdCl2 (1-100 nM) and cell proliferation was assayed using bromodeoxyuridine (BrdU) method. The mechanism underlying the proliferative effect of Cd mediated by PRs was examined using cell transfection with PR- small interfering RNA (siRNA) and western blot analysis. Our results showed the involvement of PRs in Cd induced proliferation of ovarian cancer cells. Progesterone receptors are involved in proliferative effect of Cd. Moreover, Cd modified the expression of PRA and PRB and induced ovarian cancer cell proliferation through the change of PRA/PRB ratio. In conclusion, there is a mechanistic association between Cd effects on ovarian cancer cell proliferation, estrogen receptors and PRs expression.

12.
J Cancer Res Ther ; 13(1): 107-112, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28508842

RESUMO

AIM OF THE STUDY: The cell cycle, a vital process that involves in cells' growth and division, lies at the heart of cancer. It has been shown that IB-MECA, an A3 adenosine receptor agonist inhibits the proliferation of cancer cells by inducing cell cycle arrest in several tumors. In this study, we evaluated the role of IB-MECA inhibition in cell cycle progression in ovarian cancer cells. MATERIALS AND METHODS: Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in Caov-4 and OVCAR-3. Analysis of cell cycle distribution was carried out by flow cytometry. To determine the mechanisms of IB-MECA-mediated induction of cell cycle arrest, the expression of cell cycle regulatory proteins Cyclin D1 and cyclin-dependent kinase 4 (CDK4) was evaluated. RESULTS: Our results showed that IB-MECA significantly reduced cell viability in a dose-dependent manner. Moreover, our results indicated that a low concentration of IB-MECA induced G1 cell cycle arrest. Reduction of Cyclin D1 and CDK4 protein levels was also observed after treating cancer cells with IB-MECA. CONCLUSION: This study demonstrated that IB-MECA induces G1 phase cell cycle arrest through Cyclin D1/CDK4-mediated pathway in ovarian cancer cells.


Assuntos
Agonistas do Receptor A3 de Adenosina/administração & dosagem , Adenosina/análogos & derivados , Ciclina D/genética , Quinase 4 Dependente de Ciclina/genética , Neoplasias Ovarianas/tratamento farmacológico , Adenosina/administração & dosagem , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Citometria de Fluxo , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Receptor A3 de Adenosina/genética , Transdução de Sinais/efeitos dos fármacos
13.
J Cell Biochem ; 118(9): 2909-2920, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28230290

RESUMO

Numerous studies have demonstrated the role of A3 adenosine receptor (A3AR) and signaling pathways in the multiple aspects of the tumor. However, there is a little study about the function of A3AR in the biological processes of cancer stem cells (CSCs). CSCs have a critical role in the maintenance and survival of breast cancer. The aim of current study was to investigate the effect of A3AR agonist on breast cancer stem cells (BCSCs). XTT assay showed antiproliferative effect of A3AR agonist (Cl-IB-MECA) on BCSCs. Our results also demonstrated that A3AR agonist reduces mammosphere formation in a dose-dependent manner. Flow cytometry analysis showed that A3AR agonist induces G1 cell cycle arrest and apoptosis in BCSCs. Western blot assay showed that A3AR agonist inhibits the expression of cell cycle and apoptotic regulatory proteins as well as the expression of ERK1/2 and GLI-1 proteins. Finally, these findings propose that A3AR agonist induces cell cycle arrest and apoptosis in BCSCs by inhibition of ERK1/2 and GLI-1 cascade. J. Cell. Biochem. 118: 2909-2920, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Agonistas do Receptor A3 de Adenosina/farmacologia , Neoplasias da Mama/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor A3 de Adenosina/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Receptor A3 de Adenosina/genética
14.
Cell Signal ; 32: 104-114, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28104444

RESUMO

Previous studies have shown that leptin, an adipocyte-secreted hormone, stimulates ovarian cancer invasion. Here, we investigated the contribution of uPA in leptin-induced ovarian cancer cell invasion. The cell invasion and migration experiments were carried out using matrigel invasion and wound healing assays in ovarian cancer cell lines (OVCAR3, SKOV3and CaoV-3). The mechanism underlying the invasive effect of leptin was examined using cell transfection with Ob-Rb siRNA, pre-treatment with a specific inhibitor of RhoA and ROCK, RhoA activation assay, OB-Rb, Rock and upA protein expression. Our results show that leptin induced ovarian cancer cell invasion via up-regulating upA in a time and dose-dependent manner, which was attenuated using knockdown of OB-Rb by siRNA. Moreover, pre-incubation with C3 (inhibitor of RhoA) and Y-27632 (inhibitor of ROCK) effectively attenuated leptin-induced upA expression and inhibited invasive ability of ovarian cancer cells. We also found that pretreatment with inhibitors of PI3K/AKT (LY294002), JAK/STAT (AG490) and NF-kB (BAY 11-7082) significantly reduced leptin-induced upA expression. Collectively, our findings demonstrate that OB-Rb, RhoA/ROCK, PI3K/AKT, JAK/STAT pathways and NF-kB activation are involved in leptin-induced upA expression. These results may provide a new mechanism that facilitates leptin-induced ovarian cancer invasion.


Assuntos
Leptina/farmacologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Inativação Gênica/efeitos dos fármacos , Humanos , NF-kappa B/metabolismo , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Receptores para Leptina/metabolismo
15.
Mol Med Rep ; 15(1): 467-473, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27922682

RESUMO

Active vitamin D has several antitumor effects, including prodifferentiative, antiproliferative and proapoptotic functions in a number of tissues via its binding to vitamin D receptor. The 24­hydroxylase (CYP24A1) and 1­hydroxylase (CYP27B1) enzymes are considered to be pivotal determinants of the local concentration of active vitamin D. The aim of the present study was to investigate the mRNA expression levels of the CYP24A1 and CYP27B1 genes in malignant and normal breast tissues. The tumor and adjacent normal tissue samples of 30 patients with breast cancer were acquired from the Iran National Tumor Bank, Imam Hospital (Tehran, Iran). RNA was extracted and, following cDNA synthesis, Taq­Man quantitative polymerase chain reaction analysis was performed for CYP24A1 and CYP27B1. The results demonstrated that the mRNA expression of CYP27B1 was downregulated in the tumor tissues, compared with the adjacent normal tissues (P<0.01), whereas the mRNA expression of CYP24A1 was significantly upregulated in the tumor tissues (P<0.01). This major difference revealed that the normal breast tissues transcriptionally expressed CYP24A1 slightly. These results are suggestive of dysregulation of the vitamin D signaling and metabolic pathways during tumorigenesis in breast cancer. Local alterations in the anabolism and catabolism of active vitamin D in breast cancer by the CYP27B1 and CYP24A1 may impair its anticancer functions.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Neoplasias da Mama/genética , Mama/patologia , Regulação Neoplásica da Expressão Gênica , Transcriptoma , Vitamina D3 24-Hidroxilase/genética , Adulto , Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/genética , Regulação para Cima
16.
Clin Lab ; 62(8): 1461-1468, 2016 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28164608

RESUMO

BACKGROUND: The actions of Vitamin D in different tissues, including breast tissue, are mediated by vitamin D receptor (VDR). Vitamin D has antitumor functions in the body; any changes in VDR expression can therefore affect the anticancer activities of Vitamin D. The present study was conducted to compare expression levels of VDR mRNA and protein in normal and tumor breast tissues. METHODS: Tumor and adjacent normal tissue samples from 30 patients with breast cancer were procured from the Iran National Tumor Bank of the Cancer Institute. After the extraction of RNA and cDNA synthesis, expression of the VDR gene was analyzed using Real Time RT-PCR based on TaqMan method. The expression of VDR protein was also assessed using the western blotting method. The results were quantified and analyzed in Alpha Ease, SPSS, and Excel. RESULTS: VDR mRNA and protein expression was significantly greater in tumor tissues compared to in the adjacent normal tissues (p < 0.01). Comparison of the relationship between the VDR gene mRNA expression level in tumor tissues and the clinicopathological parameters (including tumor stage, grade, size, patients' age groups, and the presence or absence of lymphatic invasion) showed VDR gene expression to be significantly related to tumor size and stage (p < 0.05). However, no relationships were observed between the expression of VDR protein in the tumor tissues and either of the parameters examined. CONCLUSIONS: The results suggest possible changes in the vitamin D signaling pathway associated with carcinogenesis of the breast, which can affect the anticancer activities of vitamin D. The study of blood vitamin D concentrations and expression changes of its anabolic and catabolic pathway enzymes can probably promote our understanding of the effects of vitamin D and its changes during breast tumorigenesis.


Assuntos
Neoplasias da Mama/metabolismo , Mama/metabolismo , Receptores de Calcitriol/genética , Adulto , Idoso , Neoplasias da Mama/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro/análise , Receptores de Calcitriol/análise , Receptores de Calcitriol/fisiologia , Vitamina D/sangue
17.
Asian Pac J Cancer Prev ; 16(7): 2601-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25854335

RESUMO

Prostate cancer, with a lifetime prevalence of one in six men, is the second cause of malignancy-related death and the most prevalent cancer in men in many countries. Nowadays, prostate cancer diagnosis is often based on the use of biomarkers, especially prostate-specific antigen (PSA) which can result in enhanced detection at earlier stage and decreasing in the number of metastatic patients. However, because of the low specificity of PSA, unnecessary biopsies and mistaken diagnoses frequently occur. Prostate cancer has various features so prognosis following diagnosis is greatly variable. There is a requirement for new prognostic biomarkers, particularly to differentiate between inactive and aggressive forms of disease, to improve clinical management of prostate cancer. Research continues into finding additional markers that may allow this goal to be attained. We here selected a group of candidate biomarkers including PSA, PSA velocity, percentage free PSA, TGFß1, AMACR, chromogranin A, IL-6, IGFBPs, PSCA, biomarkers related to cell cycle regulation, apoptosis, PTEN, androgen receptor, cellular adhesion and angiogenesis, and also prognostic biomarkers with Genomic tests for discussion. This provides an outline of biomarkers that are presently of prognostic interest in prostate cancer investigation.


Assuntos
Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/metabolismo , Antígeno Prostático Específico/sangue , Próstata/patologia , Neoplasias da Próstata/patologia , Apoptose/fisiologia , Adesão Celular/fisiologia , Humanos , Masculino , Neovascularização Patológica/patologia , Prognóstico , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/mortalidade
18.
Biochem Cell Biol ; 93(4): 321-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25877700

RESUMO

A2b adenosine receptor (A2bAR) acts as a potent regulator of cell growth in various cell lines. The present study was designed to understand the controlling mechanism of A2bAR agonist (NECA)-induced apoptosis in ovarian cancer cells. Real-time PCR and western blotting assays were used to evaluate the gene and protein expression profiles of A2bAR, respectively. MTT assay was used to study the cell proliferation effect of A2bAR agonist (NECA). Detection of apoptosis was conducted using annexin V-FITC/PI staining, caspase-3 activation assay, and the expression of Bax and Bcl-2 proteins analysis. The mitochondrial membrane potential (ΔΨM) was analyzed by employing JC-1 prob. The mRNA and protein expression levels of A2bAR in ovarian cancer cells were detected. NECA significantly reduced cell viability in a dose-dependent manner in OVCAR-3 and Caov-4 cell lines. The growth inhibition effect of NECA was related to the induction of cell apoptosis, which was manifested by annexin V-FITC staining, activation of caspase-3, and loss of mitochondrial membrane potentials (ΔΨm). In addition, downregulation of the regulatory protein Bcl-2 and upregulation of Bax protein by NECA were also observed. These findings demonstrated that NECA induces apoptosis via the mitochondrial signaling pathway. Thus, A2bAR agonists may be a potential agent for induction of apoptosis in ovarian cancer cells.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Caspase 3/metabolismo , Proliferação de Células , Neoplasias Ovarianas/patologia , Receptor A2B de Adenosina/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Potencial da Membrana Mitocondrial , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Receptor A2B de Adenosina/metabolismo
19.
Prostate ; 75(7): 735-47, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25704103

RESUMO

BACKGROUND: Adenosine, a purine nucleoside plays important roles in the pathogenesis of cancer initiation and promotion via interaction with four adenosine receptors. In the present study we examined the differential expression pattern of adenosine receptors in the malignant and adjacent normal human prostate tissues. METHODS: Prostate cancer tissue samples and adjacent normal tissues were obtained from 20 patients undergoing radical prostatectomy and histopathological diagnosis was confirmed for each sample. Total RNA was extracted and reverse transcribed into cDNA and the mRNA expression levels of adenosine receptors were investigated by Taq-man real-time RT-PCR experiment. Quantitative protein analysis was done by Western blotting experiment. Moreover, the mRNA and protein expression levels of adenosine receptors were measured after androgen treatment. RESULT: Taq-man real-time RT-PCR measurements show different expression levels of adenosine receptor transcripts. A2B adenosine receptor was predominantly expressed in tumor tissues (2.4-fold) followed by significantly expression of A3 (1.6-fold) and A2A adenosine receptors (1.5-fold) compared to adjacent normal tissues. The presence of adenosine receptors at protein levels in prostate cancer tissues compared with normal tissues was shown the following rank order: A2B > A3 > A2A > A1 . Androgen receptor regulates adenosine receptors mRNA and protein expression in AR-positive LNCaP cells, which was not seen in AR-negative PC-3 cells. CONCLUSION: These results indicated for the first time, the differential mRNA expression profile and protein levels of adenosine receptors in the human prostate cancer. Interestingly, the A2B adenosine receptor followed by A3 is highly expressed in prostate tumor samples in comparison with the adjacent normal tissues. The findings support the possible key role of A2B adenosine receptor in promoting cancer cell growth and suggest that A2B may be a novel target for prostate cancer treatment.


Assuntos
Neoplasias da Próstata/metabolismo , Receptores Purinérgicos P1/biossíntese , Idoso , Western Blotting , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Purinérgicos P1/classificação , Receptores Purinérgicos P1/genética
20.
Toxicol Mech Methods ; 25(1): 48-55, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25403798

RESUMO

CONTEXT AND OBJECTIVE: Although low concentrations of cadmium exposure may enhance growth of human cultured cells, high and long term of this heavy metal leads to cell death through apoptosis or necrosis. This study was conducted to define the underlying biochemical mechanism of Cd-induced cell death in MCF-7 human breast cancer cell line. METHODS: The MCF-7 breast cancer cells were treated with different concentrations of CdCl2 and cell viability was assessed using the MTT assay. A propidium iodide (PI) and annexin-V staining flow cytometric method was used for apoptosis detection. Hoechst 33342 staining was used to observe the morphological changes of cell apoptosis. The cellular DNA was isolated using DNA kit extraction and analyzed electrophoretically. Intracellular reactive oxygen species (ROS) levels were quantified using the fluorescent dye (DCFH-DA). RESULTS: A progressive loss in cell viability and an increased number of apoptotic cells were observed upon 48 h exposure to CdCl2. N-acetylcysteine (NAC) administration reversed the cadmium cytotoxicity effects and protected cells from apoptotic death. Simultaneously, significant elevations of ROS levels were revealed in a dose-dependent manner during the exposure. Typical morphological changes of apoptosis were observed with Hoechst staining after cadmium treatment. CONCLUSION: These results suggest that during the apoptosis mediated by cadmium chloride, ROS production and oxidative damage may be an initiating event and responsible for the mechanism of MCF-7 human breast cell death.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cloreto de Cádmio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/farmacologia , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA , Relação Dose-Resposta a Droga , Feminino , Humanos , Células MCF-7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...