Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
País como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell Physiol Biochem ; 57(2): 105-122, 2023 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-37052042

RESUMO

BACKGROUND/AIMS: Macrophages interact with tumor cells within the tumor microenvironment (TME), which plays a crucial role in tumor progression. Cancer cells also can instruct macrophages to facilitate the spread of cancer and the growth of tumors. Thus, modulating macrophages-cancer cells interaction in the TME may be therapeutically beneficial. Although calcitriol (an active form of vitamin D) has anticancer properties, its role in TME is unclear. This study examined the role of calcitriol in the regulation of macrophages and cancer cells in the TME and its influence on the proliferation of breast cancer cells. METHODS: We modeled the TME, in vitro, by collecting conditioned medium from cancer cells (CCM) and macrophages (MCM) and culturing each cell type separately with and without (control) a high-dose (0.5 µM) calcitriol (an active form of vitamin D). An MTT assay was used to examine cell viability. Apoptosis was detected using FITC (fluorescein isothiocyanate) annexin V apoptosis detection kit. Western blotting was used to separate and identify proteins. Quantitative real-time PCR was used to analyze gene expression. Molecular docking studies were performed to evaluate the binding type and interactions of calcitriol to the GLUT1 and mTORC1 ligand-binding sites. RESULTS: Calcitriol treatment suppressed the expression of genes and proteins implicated in glycolysis (GLUT1, HKII, LDHA), promoted cancer cell apoptosis, and reduced viability and Cyclin D1gene expression in MCM-induced breast cancer cells. Additionally, calcitriol treatment suppressed mTOR activation in MCM-induced breast cancer cells. Molecular docking studies further showed efficient binding of calcitriol with GLUT1 and mTORC1. Calcitriol also inhibited CCM-mediated induction of CD206 and increased TNFα gene expression in THP1-derived macrophages. CONCLUSION: The results suggest that calcitriol may impact breast cancer progression by inhibiting glycolysis and M2 macrophage polarization via regulating mTOR activation in the TME and warrants further investigation in vivo.


Assuntos
Neoplasias da Mama , Calcitriol , Humanos , Feminino , Calcitriol/farmacologia , Calcitriol/uso terapêutico , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Simulação de Acoplamento Molecular , Microambiente Tumoral/genética , Serina-Treonina Quinases TOR/metabolismo , Macrófagos/metabolismo , Neoplasias da Mama/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Glicólise , Proliferação de Células/genética , Linhagem Celular Tumoral , Ativação de Macrófagos
2.
Int J Mol Sci ; 22(12)2021 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-34207035

RESUMO

Breast cancer is the most commonly occurring cancer in women of Western countries and is the leading cause of cancer-related mortality. The breast tumor microenvironment contains immune cells, fibroblasts, adipocytes, mesenchymal stem cells, and extracellular matrix. Among these cells, macrophages or tumor-associated macrophages (TAMs) are the major components of the breast cancer microenvironment. TAMs facilitate metastasis of the breast tumor and are responsible for poor clinical outcomes. High TAM density was also found liable for the poor prognosis of breast cancer. These observations make altering TAM function a potential therapeutic target to treat breast cancer. The present review summarizes the origin of TAMs, mechanisms of macrophage recruitment and polarization in the tumor, and the contributions of TAMs in tumor progression. We have also discussed our current knowledge about TAM-targeted therapies and the roles of miRNAs and exosomes in re-educating TAM function.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Microambiente Tumoral , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Animais , Biomarcadores Tumorais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Comunicação Celular , Progressão da Doença , Suscetibilidade a Doenças , Exossomos/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Imunomodulação , Ativação de Macrófagos/imunologia , MicroRNAs/genética , Metástase Neoplásica , Estadiamento de Neoplasias , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Carga Tumoral , Macrófagos Associados a Tumor/patologia
3.
Clin Exp Pharmacol Physiol ; 42(5): 451-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25800075

RESUMO

The extent to which cytochrome P450 (CYP) 2C19 genotype influences the effectiveness of clopidogrel remains uncertain due to considerable heterogeneity between studies. We used the polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) method for genotyping loss of function (LOF) allele, CYP2C19*2 and gain of function (GOF) allele, CYP2C19*17 in 163 patients undergoing PCI and 165 healthy volunteers from an ethnically distinctive Bangladeshi population. Thirty-eight patients took prasugrel and 125 patients took clopidogrel among whom 30 patients had their clopidogrel active metabolites (CAM) determined by LC-MS/MS 1-1.5 h after clopidogrel intake. All patients who underwent PCI had their P2Y12 per cent inhibition (PRI) measured by VerifyNow System. The impact of different genotypes on CAM and PRI were also determined. We did not find significant variation of CYP2C19*2 (P > 0.05) and CYP2C9*17 (P > 0.05) alleles among healthy volunteers and patients. CAM concentration as well as PRI by clopidogrel varied significantly (P < 0.05) based on genotypic variation of CYP2C19*2 and CYP2C19*17 individually. Such influence was not observed in case of prasugrel. Genotypic variation did not impact PRI but as a whole PRI by prasugrel was better than that of clopidogrel (P < 0.05). Due to presence of both of alleles the effect on PRI by clopidogrel could not be predicted, effectively indicating possible involvement of other factors. Genotype guided clopidogrel dose adjustment would be beneficial and therefore we propose mandatory genotyping before clopidogrel dosing. Prasugrel proved to be less affected by genotypic variability, but due to lack of sufficient long-term toxicity data, caution would be adopted before substituting clopidogrel.


Assuntos
Citocromo P-450 CYP2C19/genética , Frequência do Gene , Cloridrato de Prasugrel/farmacologia , Cloridrato de Prasugrel/farmacocinética , Ticlopidina/análogos & derivados , Bangladesh , Clopidogrel , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Intervenção Coronária Percutânea , Ticlopidina/farmacocinética , Ticlopidina/farmacologia
4.
Life Sci ; 301: 120610, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35525305

RESUMO

AIMS: Breast cancer metastasis is the leading cause of mortality among breast cancer patients. Epithelial to mesenchymal transition (EMT) is a biological process that plays a fundamental role in facilitating breast cancer metastasis. The present study assessed the efficacy of parthenolide (PTL Tanacetum parthenium) on EMT and its underlying mechanisms in both lowly metastatic, estrogen-receptor positive, MCF-7 cells and highly metastatic, triple-negative MDA-MB-231 cells. MAIN METHODS: MCF-7 and MDA-MB-231 cells were treated with PTL (2 µM and 5 µM). Cell viability was determined by MTT (3-(4,5-dimethy lthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay. Apoptosis was analyzed by the FITC (fluorescein isothiocyanate) annexin V apoptosis detection kit. The monolayer wound scratch assay was employed to evaluate cancer cell migration. Proteins were separated and identified by Western blotting. Gene expression was analyzed by quantitative real-time PCR. KEY FINDINGS: PTL treatment significantly reduced cell viability and migration while inducing apoptosis in both cell lines. Also, PTL treatment reverses the EMT process by decreasing the mesenchymal marker vimentin and increasing the epithelial marker E-cadherin compared to the control treatment. Importantly, PTL downregulates TWIST1 (a transcription factor and regulator of EMT) gene expression, concomitant with the reduction of transforming growth factor beta1 (TGFß1) protein and gene expression in both cell lines. Additionally, molecular docking studies suggest that PTL may induce anticancer properties by targeting TGFß1 in both breast cancer cell lines. SIGNIFICANCE: Our findings provide insights into the therapeutic potential of PTL to mitigate EMT and breast cancer metastasis. These promising results demand in vivo studies.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Simulação de Acoplamento Molecular , Sesquiterpenos , Fator de Crescimento Transformador beta1/metabolismo
5.
J Nutr Biochem ; 53: 111-120, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29216499

RESUMO

Breast cancer is one of the major causes of death in the USA. Cancer cells, including breast, have high glycolysis rates to meet their energy demands for survival and growth. Vitamin D3 (VD3) is important for many important physiological processes such as bone mineralization, but its anticancer role is yet to be proven. We find that VD3 treatment significantly down-regulates glycolytic enzymes and genes and decreases glucose uptake - for both lowly metastatic MCF-7 and highly metastatic MDA-MB-231 (MB231) breast cancer cells. VD3 also significantly decreases cell viability by inducing apoptosis - consistent with decreased expression of mammalian target of rapamycin (mTOR), which regulates glycolysis and cancer cell survival, and increases 5' adenosine monophosphate-activated protein kinase (AMPK) activation. These changes accompany a significant reduction of cell migration and increased cell stiffness, presumably a consequence of reversal of the epithelial to mesenchymal transition resulting in increased E-cadherin, and F-actin, and reduced vimentin expression. High levels of cytoskeletal and cortical F-actin may cause high cell stiffness. VD3-induced mechanical changes are stronger in highly metastatic MB231 than in lowly metastatic MCF-7 cells. Our results suggest therapeutic and preventive roles of VD3 in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Colecalciferol/farmacologia , Glicólise/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Enzimas/genética , Enzimas/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacocinética , Glicólise/fisiologia , Humanos , Ácido Láctico/metabolismo , Células MCF-7 , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa