Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
J Cell Mol Med ; 28(19): e70132, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39350724

RESUMEN

Aging is a risk factor for various human disorders, including cancer. Current literature advocates that the primary principles of aging depend on the endogenous stress-induced DNA damage caused by reactive oxygen species 50 Hz low-frequency magnetic field was suggested to induce DNA damage and chromosomal instability. NF-kB, activated by DNA damage, is upregulated in age-related cancers and inhibition of NF-kB results in aging-related delayed pathologies. Metformin (Met), an NF-kB inhibitor, significantly reduces both NF-kB activation and expression in aging and cancer. This in vitro study, therefore, was set out to assess the effects of 5mT MF in 50 Hz frequency and Met treatment on the viability and proliferation of aged mouse NIH/3T3 fibroblasts and expression of RELA/p65, matrix metalloproteinases MMP2 and MMP9, and E-cadherin (CDH1) genes. The trypan blue exclusion assay was used to determine cell viability and the BrdU incorporation assay to determine cell proliferation. The MMP-2/9 protein analysis was carried out by immunocytochemistry, NF-kB activity by ELISA and the expressions of targeted genes by qRT-PCR methods. Four doses of Met (500 uM, 1 mM, 2 mM and 10 mM) suppressed both the proliferation and viability of fibroblasts exposed to the MF in a dose-dependent pattern, and the peak inhibition was recorded at the 10 mM dose. Met reduced the expression of NF-kB, and MMP2/9, elevated CDH1 expression and suppressed NF-kB activity. These findings suggest that Met treatment suppresses the carcinogenic potential of 50 Hz MFs in aged mouse fibroblasts, possibly through modulation of NF-kB activation and epithelial-mesenchymal transition modulation.


Asunto(s)
Proliferación Celular , Supervivencia Celular , Fibroblastos , Campos Magnéticos , Metformina , FN-kappa B , Animales , Metformina/farmacología , Ratones , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Células 3T3 NIH , FN-kappa B/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/patología , Factor de Transcripción ReIA/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Cadherinas/metabolismo , Cadherinas/genética , Senescencia Celular/efectos de los fármacos
2.
Eur J Breast Health ; 20(3): 167-177, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-39257008

RESUMEN

Midkine (MDK) is a protein that contributes to both physiological and pathological processes. Several studies provide insight into the different roles of MDK in development, tissue repair, neural plasticity, and health and disease processes. This research further examined how MDK contributed to conditions, including neurological diseases, inflammation, and ischaemia. Furthermore, MDK overexpression has been reported in many kinds of cancer and MDK is recognized as a malignancy marker. MDK stimulates pro-tumor activity by regulating a number of signaling pathways, which increase cancer cell proliferation, survival, metastasis, and treatment resistance. However, studies have shown that MDK also functions as a molecule that regulates drug resistance. Several cancer therapy techniques have been suggested to modify MDK function, including antibody-based therapies, oligonucleotides, oncolytic viruses, and small compounds. Further research and experimentation will be required to establish the therapeutic relevance and efficacy of these treatments. This review focuses on the role of MDK in cancer biology, as well as its multiple different roles in health and disease processes.

3.
Med Hypotheses ; 134: 109420, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31634770

RESUMEN

Metformin, a drug widely used in the treatment of type II diabetes mellitus (T2DM), has been the focus of interest as a potential therapeutic agent for certain types of malignancies, including gynaecological cancers [i.e. endometrial cancer (EC)]. Although the exact mechanism behind the potential anticancer activity of metformin is still not completely understood, certain studies have suggested that different effects on cell functions, such as inhibition of cell migration, apoptosis and tumor cell proliferation, are involved in its preventive and therapeutic effects in certain types of malignancies, including EC. In contrast, midkine (MK), a heparin-binding growth factor and cytokine, which induces carcinogenesis and chemoresistance, promotes the development and progression of many malignant tumours by increasing diverse cell functions such as cell proliferation, cell survival and antiapoptotic activities via mainly the activation of phosphatidyl inositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. The same pathways are also subject to certain therapeutic effects of metformin, although this cytokine and this drug have some different mechanism of action pathways as well. Taken together, MK and metformin appear to have opposite effects in various biological processes such as apoptosis, cell proliferation, cell survival, cell migration, and angiogenesis. On the other hand, MK activates PI3K and MAPK cell signal pathways, whereas metformin inhibits these two pathways. It seems likely that almost all the pathways and cell functions, which play important roles in malignancies, are inhibited by metformin and activated by MK. Given the opposite relationship between the actions of metformin and MK, we hypothesize that metformin may act like a novel MK inhibitor in some malignancies. We also discuss the possible relationship between metformin and MK in the context of EC, the most common gynecological cancer worldwide, which incidence is rising rapidly, in parallel with the increase in obesity, T2DM and insulin resistance. In this respect, the therapeutic use of metformin may improve the survival of EC or other cancers, via inhibiting or overcoming the unwanted effects of MK in carcinogenesis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/prevención & control , Metformina/farmacología , Midkina/antagonistas & inhibidores , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Fenómenos Fisiológicos Celulares/efectos de los fármacos , Reposicionamiento de Medicamentos , Neoplasias Endometriales/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Humanos , Hipoglucemiantes/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Integrinas/fisiología , Metformina/uso terapéutico , Midkina/fisiología , Modelos Biológicos , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
4.
Invest New Drugs ; 38(2): 246-263, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-30993586

RESUMEN

Prostate cancer (PCa) is the most common cancer in men worldwide. Midkine (MK) is overexpressed in PCa, as well as in tumor-initiating cells termed cancer stem cells (CSCs). Apigenin is a dietary flavone with considerable anti-tumor activities. In this study, we explored the possible therapeutic use of MK silencing, apigenin treatment, and a combination of both on human PCa and prostate cancer stem cells (PCSCs). CD44+CD133+ PC3 and CD44+ LNCaP CSCs were isolated from their parent cell lines. Both MK knockdown and apigenin treatment resulted in loss of cell viability in PCSCs, and these effects were significantly elevated when apigenin was applied with MK silencing. Combined treatment of CD44+CD133+ PC3 cells with apigenin and MK siRNA was also more effective in inducing apoptotic and non-apoptotic cell death when compared with individual applications. Treatment of CD44+ LNCaP cells with apigenin significantly decreased viability, although the combination treatment did not markedly alter the individual therapy. Molecular events underlying cell cycle arrest and inhibition of the survival, proliferation, and migration of CD44+CD133+ PC3 cells were found to be associated with upregulated p21, p27, Bax, Bid, caspase-3, and caspase-8 expression, as well as downregulated p-p38, p-ERK, NF-κB, and PARP. In addition, the combination of apigenin treatment and MK silencing showed better outcomes on the anticancer efficacy of docetaxel in CD44+CD133+ PC3 cells. In conclusion, MK-regulated events are different between PCSCs, and when combined with apigenin plus MK silencing, docetaxel treatment may be a valuable approach for the eradication of PCSCs.


Asunto(s)
Antineoplásicos/farmacología , Apigenina/farmacología , Docetaxel/farmacología , Midkina/genética , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/terapia , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Silenciador del Gen , Humanos , Masculino , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Crit Rev Oncog ; 24(1): 35-45, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31679218

RESUMEN

Midkine (MK) is a heparin-binding anti-apoptotic growth factor or cytokine also known as neurite growth-promoting factor 2 (NEGF2). It is developmentally an important retinoic acid-responsive gene product strongly induced during the mid-gestation stage. Midkine promotes different cellular events such as cell growth, differentiation, survival, gene expression, and drug resistence. Midkine, the phosphatidylinositol 3-kinases (PI3-kinase, PI3K) and glycogen synthase kinase-3 beta (GSK-3ß) inhibitors together with lithium chloride may be a very effective treatment modality, especially in tumors with high expression of these two molecules. PI3 kinase and GSK-3ß, both serine threonine kinases located in the center of the signaling network, are very important regulator molecules for cell survival or death. Lithium chloride (LiCl), with its newly discovered antineoplastic effect and cytotoxicity potentiation, has become a promising agent in the application of new combination treatments. Although the LiCl mechanism of action is still not fully understood, previous studies have shown that LiCl is an inhibitör of the inositol monophosphatase (IMPase) and GSK-3ß. GSK-3ß, is a serine-threonine protein kinase involved in cell proliferation, differentiation, survival, apotosis, and tumorogenesis. The role of GSK-3ß in tumorigenesis and cancer remains controversial. It may have a function as a tumor suppressor for certain types of tumors, but it promotes cell growth and development in other tumor types.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cloruro de Litio/farmacología , Cloruro de Litio/uso terapéutico , Midkina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Biomarcadores , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Midkina/antagonistas & inhibidores , Midkina/química , Midkina/genética , Neoplasias/etiología , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
6.
Tissue Cell ; 59: 18-32, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31383285

RESUMEN

OBJECTIVES: To define ultrastructural features accompanying to antitumor effects of gemcitabine, vinorelbine and cyclooxygenase inhibitors in C6 glioma cells in vitro. Vinorelbine is a semisynthetic vinca alkaloid and recent studies showed its antitumor activity in pediatric optic and pontine gliomas. Vinorelbine infusion induces a severe tumor site-pain in systemic cancers, but it is unknown whether algesia and inflammation contribute to its antitumor effects. Gemcitabine is a nucleoside-chemotherapeutic which was recently shown to act as a radiosensitizer in high-grade glioma. Some studies showed synergism of anti-inflammatory cyclooxygenase-inhibitors with microtubule inhibitors and gemcitabine. DMSO is a solvent and blocks both cylooxygenase and ribonucleotide reductase, another target of gemcitabine. Rofecoxib is withdrawn from the market, yet we used it for investigational purposes, since it blocks cylooxygenase-2 1000-times more potently than cylooxygenase -1 and is also a selective inhibitor of crinophagy. METHODS: Plating efficacy, 3D-spheroid S-phase analysis with BrdU labelling and transmission electron microscopical analyses were performed. RESULTS: Vinorelbine induced frequent mitotic slippage/apoptosis and autophagy. Despite both DMSO and rofecoxib induced autophagy alone and in synergy, they reduced mitotic catastrophe and autophagy triggered by vinorelbine, which was also reflected by reduced inhibition of spheroid S-phase. Gemcitabine induced karyolysis and margination of coarse chromatin towards the nuclear membrane, abundant autophagy, gutta adipis formation and decrease in mitochondria, which were enhanced by DMSO and rofecoxib. CONCLUSIONS: Detailed ultrastructural analysis of the effects of chemotherapeutic drugs may provide a broader insight about their actions and pave to develop better strategies in treatment of glioblastoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Muerte Celular Autofágica/efectos de los fármacos , Glioblastoma , Inhibidores de la Ciclooxigenasa/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/ultraestructura , Humanos , Vinorelbina/farmacología , Gemcitabina
7.
Biomed Res Int ; 2019: 1407167, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30792990

RESUMEN

Epigenetic changes have major role in the normal development and programming of gene expression. Aberrant methylation results in carcinogenesis. The primary objective of our study is to determine whether primary tumor tissue and cultured tumor cells in 2D and 3D tissue culture systems have the same methylation signature for PAX5, TMPRSS2, and SBDS. These findings will play an important role in developing in vitro model system to understand the effect of methylation inhibitors on primary tumor tissue. In a previous study PAX5, TMPRSS2, and SBDS genes that we are investigating were reported to be methylated more than 60% in breast cancer and malignant melanoma cell lines. However, these genes have never been studied in primary tumor tissues. Thus, primary tumor tissues of breast cancer and malignant melanoma were first grown in 2D and 3D cultures. Then these two types of tumor tissues and their 2D and 3D cultures were investigated for changes considering methylation levels in PAX5, TMPRSS2, and SBDS genes using real-time polymerase chain reaction. No differences were observed in the primary tissues and culture systems for both PAX5 and TMPRSS2 in malignant melanoma tissues. We found that PAX5 gene was an efficient marker to measure the effects of methylation inhibitors for in vitro systems for malignant melanoma tissue.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Metilación de ADN/genética , Melanoma/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/patología , Neoplasias Primarias Desconocidas , Regiones Promotoras Genéticas/genética , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Técnicas de Cultivo de Tejidos
8.
Clin Neurol Neurosurg ; 177: 77-85, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30622045

RESUMEN

OBJECTIVE: Risk of high grade gliomas is lower in young females and its incidence enhances after menopause suggesting likely protective roles of female hormones. Hormone replacement therapy (HRT) was widely employed to treat osteoporosis and some epidemiological studies showed that HRT regimes including progesterone analogs such as medroxyprogesterone acetate (MPA) decreased risk of glial tumors. Tibolone is a unique progesterone analog employed in HRT with tissue specific estrogenic effects and stimulates gene expressions very similar to those induced by MPA. Tibolone's pro-estrogenic effects occur particularly in bone and brain and both MPA and tibolone inhibit AKR1C enzymes, which involve in temozolomide chemoresistance. Hence, we aimed to investigate interactions between MPA, tibolone and temozolomide in modification of glioma cell growth and fine structure. PATIENTS AND METHODS: For our studies, we have particularly chosen C6 rat glioma cell line due to several reasons: i) We previously showed that MPA reduced growth and induced procarbazine-sensitization in C6 cells; ii) temozolomide has a triazene-type molecular structure like procarbazine; iii) other groups previously showed that C6 glioma cell line is more resistant to temozolomide than human glioma cells; hence it may provide a native model of chemoresistance. Monolayer plating efficacy, soft agar colony growth, 3D-spheroid S-phase (as determined by BrdU-labeling) and electron microscopical analyses were performed to assess mutual interactions between MPA, tibolone and temozolomide. RESULTS: MPA inhibited clonogenic growth of C6 glioma and this effect is augmented by both tibolone and temozolomide. MPA and tibolone inhibited DNA synthesis in C6 glioma spheroids to similar levels which can be achieved with temozolomide. Electron microscopical analyses revealed synergisms between MPA, tibolone and temozolomide involved mitochondrial proliferation, condensation, mitophagy and autophagy. CONCLUSIONS: MPA and tibolone shall be studied in further experimental models of glioblastoma in vitro and in vivo.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Medroxiprogesterona/farmacología , Neoplasias Encefálicas/patología , Glioblastoma/tratamiento farmacológico , Glioma/patología , Humanos , Acetato de Medroxiprogesterona/farmacología , Microscopía Electrónica/métodos , Mitocondrias/efectos de los fármacos , Temozolomida/farmacología
9.
Mod Rheumatol ; 29(4): 567-571, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30217121

RESUMEN

Midkine (MK) is a heparin-binding growth factor that markedly expressed during embryogenesis but downregulated to inconsiderable levels in healthy adults. However, MK is upregulated during tissue repair and in many pathologic conditions, mostly malignancies and inflammatory diseases. MK promotes a number of functions in target cells such as migration, proliferation, survival, growth, reproduction and repair, angiogenesis, and gene expression. It acts as a pro-inflammatory cytokine and contributes to chronic inflammation via promoting chemotaxis and tissue infiltration of neutrophils and macrophages. Furthermore, MK upregulated the production of various inflammatory mediators (i.e. interleukin (IL) 6 and IL8). Recent studies have demonstrated strong evidence that MK is involved in the onset and progression of autoimmune rheumatic diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and Sjögren's syndrome (SS) and other autoimmune conditions such as multiple sclerosis (MS). Additionally, it has been shown that MK is overexpressed in two major clinically defined forms of inflammatory bowel diseases (IBD), Crohn's disease (CD) and ulcerative colitis (UC), which are classified as autoinflammatory diseases. Taken together, MK is involved in the pathogenesis of autoimmune and autoinflammatory diseases and may serve as an indicator and biomarker in these conditions. Furthermore, MK inhibitors are expected to contribute in the management of these diseases.


Asunto(s)
Artritis Reumatoide/etiología , Lupus Eritematoso Sistémico/etiología , Midkina/metabolismo , Síndrome de Sjögren/etiología , Artritis Reumatoide/inmunología , Humanos , Lupus Eritematoso Sistémico/inmunología , Síndrome de Sjögren/inmunología
10.
Chem Biol Interact ; 294: 107-117, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30142312

RESUMEN

BACKGROUND: PPAR-δ is a transcription factor which has crucial roles in stimulating oligodendroglial differentiation and myelination and its activation was also shown to differentiate malignant C6 glioma cells into oligodendrocytes. OBJECTIVE: One of the ligands of PPAR-δ is erucic acid (EA), an edible omega-9 fatty acid consumed more by Asian populations and exists highly in Chinese womens milk. There exist epidemiological evidence that pediatric brain tumor incidence is among the lowest in the Chinese population. EA is also an ingredient of Lorenzo's oil used against adrenoleukodystrophy, a pediatric demyelinating disease. EA was inappropriately assumed as a strong cardiotoxin based on Spanish oil syndrome, caused by toxic-aniline dye refined rapeseed oil. In this study, we studied whether EA is capable to block growth of C6 glioma cells and modify cardiotoxicity of doxorubicin. MATERIALS AND METHODS: We studied effects of EA on the 3-dimensional appearance of the adherent cells, soft agar colony formation and S-phase in the 3-dimensional spheroids in C6 glioma cell cultures. We also investigated the effects of EA on hepatic and cardiac toxicity of doxorubicin. RESULTS: EA decreased in vitro growth of C6 glioma cells at therapeutically achievable concentrations. EA effects were more prominent in 3D-assays (soft agar colonies and spheroids) and induced cell fusions in monolayer cultures. EA decreased S-phase inhibitory potency of doxorubicin (DOX), yet augmented its efficacy to induce a senescent morphology (as assessed by scanning electron microscopy) in monolayer and to increase iNOS and eNOS expression in spheroids. In our study, EA reduced DOX-induced necrosis in mice heart and liver and induced healthier morphology of heart mitochondria (as assessed by transmission electron microscopy); yet intercalated disks (ID) were more disturbed with DOX + EA. CONCLUSIONS: Both the antitumor and cardiac effects of EA may associate with the cell-to-cell contact mechanisms. Combining systemic EA with intrathecal DOX-chemotherapy via Ommaya reservoirs may reduce DOX concentrations in systemic circulation, hinder toxic interactions with EA and induce selective kill of glioma cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Doxorrubicina/toxicidad , Ácidos Erucicos/farmacología , PPAR delta/metabolismo , Trioleína/química , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Combinación de Medicamentos , Ácidos Erucicos/química , Glioma/metabolismo , Glioma/patología , Corazón/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR delta/química , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
11.
Biomed Pharmacother ; 107: 793-805, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30142541

RESUMEN

AIMS: To examine the functions of growth factor midkine (MK) and a flavonoid quercetin on survival, apoptosis and migration of prostate cancer (PCa) stem cells (CSCs). MAIN METHODS: CD44+/CD133+ and CD44+ stem cells were isolated from PC3 and LNCaP cells, respectively by magnetic-activated cell sorting system. 3D cell culture was used to evaluate the ability of quercetin, MK siRNA, and the combination of both to inhibit spheroid formation, apoptosis and cell cycle arrest. Image-based cytometer, RT-qPCR, Western blotting and transwell migration assays were performed. KEY FINDINGS: Quercetin treatment for 24-72 h inhibited PC3 and CD44+/CD133+ stem cell proliferation in a time- and dose-dependent manner. Knockdown of endogenous MK expression significantly suppressed proliferation of CD44+/CD133+ and CD44+ cells as well as their parent cells. Co-administration of MK siRNA and quercetin reduced the cell survival, induced apoptosis and caused G1 phase cell cycle arrest more effectively than the individual therapy. Knockdown of MK significantly enhanced the inhibitory effect of quercetin on CD44+/CD133+ migration and spheroid formation. In addition, the combined therapy inhibited the phosphorylation of PI3K, AKT and ERK1/2, and reduced the protein expression of p38, ABCG2 and NF-κB. SIGNIFICANCE: Quercetin alone exhibited significant cytotoxic effects on CD44+/CD133+. MK plays an important role in the proliferation of CD44+/CD133+ and CD44+ cells in particular, and quercetin and MK-silencing therapy may be an important strategy in targeting CSCs that play a role in relapse, migration and drug resistance.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Midkina/farmacología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/genética , Separación Celular , Supervivencia Celular/efectos de los fármacos , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
12.
Turk J Med Sci ; 48(2): 441-448, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29714468

RESUMEN

Background/aim: In this study, the effects of resveratrol as a natural polyphenol compound, gemcitabine as an antimetabolite that has nucleoside structure analogous to deoxycytidine, and para-aminophenol-derived paracetamol were investigated with single and combined applications in monolayers of the MDAH-2774 human ovarian cancer cell line. Materials and methods: Drugs were evaluated in cell culture with respect to cell proliferation, cell cytotoxicity (trypan blue dye exclusion test), synthesis phase of cell cycle, and cell structure in 24, 48, 72, and 96 h. Result: Resveratrol and gemcitabine diminished both cell proliferation and cell cycle synthesis phase indication in monolayer cell cultures (P < 0.05). All combination groups showed similar effects that were mainly more effective in respect to single usage of resveratrol and gemcitabine in monolayer cell cultures. Conclusion: The effects of gemcitabine, resveratrol, and paracetamol were investigated in monolayers of the MDAH-2774 human ovarian cancer cell line and a decrease in cell number in cell cycle synthesis phase, prevention of cell proliferation, and destruction of cell structure were observed.

13.
Med Hypotheses ; 107: 26-28, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28915956

RESUMEN

Multidrug resistance (MDR) to chemotherapy may significantly affect the outcome of cancer treatment. ATP-dependent drug efflux pumps, including P-glycoprotein (P-gp), contribute to the resistance of various chemotherapeutic agents. Overexpression of P-gp in tumor cells induces chemoresistance via pumping the anticancer drugs out of the cells. In addition to taking part in many biological processes such as development, reproduction and repair, midkine (MK) also plays important roles in the pathogenesis of malignant diseases as well as in the regulation of MDR. Although, the mechanisms of action of P-gp and MK are different, overexpression of both proteins prevents the accumulation of many chemotherapeutics in tumor cells, leading to decreased therapeutic effects of anticancer drugs. Therefore, identification of the result of dual inhibition of P-gp and MK in overcoming chemoresistance may enhance the likelihood for a more efficient chemotherapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Midkina , Modelos Biológicos , Neoplasias/genética , Proteínas/antagonistas & inhibidores
14.
Oncol Lett ; 12(4): 2948-2955, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27703531

RESUMEN

Prostate cancer (PCa) is the second most frequent type of cancer in men worldwide and the levels of differentiation growth factor midkine (MK) are increased in PCa. Cancer and/or the treatment process itself may lead to psychiatric disorders. Lithium chloride (LiCl) has anti-manic properties and has been used in cancer therapy; however, it has a queried safety profile. In addition, cancer stem cells are responsible for the heterogeneous phenotype of tumor cells; they are involved in progression, metastasis, recurrence and therapy resistance in various cancer types. The aims of the present study were to investigate the effect of different concentrations of LiCl on PCa stem cells (whether a shift from tumorigenic to non-tumorigenic cells occurs) and to determine if these results can be explained through changes in MK levels. Monolayer and spheroid cultures of human prostate stem cells and non-stem cells were incubated with low (1, 10 µM) and high (100, 500 µM) concentrations of LiCl for 72 h. Cell proliferation, apoptotic indices, MK levels and ultrastructure were evaluated. Cells stimulated with low concentrations showed high proliferation, low apoptotic indices, high MK levels and more healthy ultrastructure. Opposite results were obtained at high concentrations. Furthermore, stem cells were more sensitive to stimulation and more resistant to inhibition than non-stem cells. LiCl exhibited concentration-dependent effects on stem cell and non-stem cell groups. MK levels were not involved in the biphasic effect of LiCl; however, they were proportionally affected. To the best of our knowledge, the present study was the first to show the effect of LiCl on PCa stem cells through MK.

15.
Life Sci ; 162: 77-86, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27569589

RESUMEN

AIMS: Cancer stem cells (CSCs) are involved in drug resistance, metastasis and recurrence of cancers. The efficacy of apigenin on cell survival, apoptosis, migration and stemness properties were analyzed in CSCs. MAIN METHODS: Prostate CSCs (CD44(+)) were isolated from human prostate cancer (PCa) PC3 cells using a magnetic-activated cell sorting system. PC3 and CSCs were treated with various concentrations of apigenin, docetaxel and their combinations for 48h. KEY FINDINGS: Apigenin dose dependently inhibited CSCs and PC3 cell survival, and this was accompanied with a significant increase of p21 and p27. Apigenin induced apoptosis via an extrinsic caspase-dependent pathway by upregulating the mRNA expressions of caspases-8, -3 and TNF-α, but failed to regulate the intrinsic pathway as determined by the Bax, cytochrome c (Cyt-c) and APAF-1 in CSCs. In contrary to CSCs, apigenin induced intrinsic apoptosis pathway as evidenced by the induction of Bax, Cyt-c and caspase-3 while caspase-8, TNF-α and Bcl-2 levels remained unchanged in PC3 cells. The flavonoid strongly suppressed the migration rate of CSCs compared to untreated cells. Significant downregulation of matrix metallopeptidases-2, -9, Snail and Slug exhibits the ability of apigenin treatment to suppress invasion. The expressions of NF-κB p105/p50, PI3K, Akt and the phosphorylation of pAkt were decreased after apigenin treatment. Moreover, apigenin treatment significantly reduced pluripotency marker Oct3/4 protein expression which might be associated with the down-regulation of PI3K/Akt/NF-κB signaling. SIGNIFICANCE: Our data indicated that, apigenin could be a useful compound to prevent proliferation and migration of cancer cells as well as CSCs.


Asunto(s)
Apigenina/farmacología , Receptores de Hialuranos/inmunología , FN-kappa B/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Apoptosis , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular , Humanos , Masculino , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología
16.
Neurol Res ; 38(9): 766-74, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27367429

RESUMEN

OBJECTIVES: Glioblastoma (GBM), the most common primary tumour of the central nervous system, is characterised by a high malignancy and poor prognosis. The aims of this study were to investigate whether the combination of imatinib mesylate (IM) and lithium chloride (LiCl) exhibited a synergistic effect in treatment and to determine whether midkine (MK) affected the fate of this treatment in vitro. METHODS: Monolayer and spheroid cultures of the T98G human GBM cell line were treated with an IM and LiCl combination for 72 h. The cell proliferation index, apoptotic index, cell cycle distribution, apoptotic and anti-apoptotic protein levels, and cAMP level as well as the cellular morphology and ultrastructure were evaluated. RESULTS: All applications inhibited cell proliferation and induced apoptosis. The most substantial decreases in cell proliferation and the caspase-3, epidermal growth factor receptor (EGFR), platelet derived growth factor receptor-alpha (PDGFR-α), multidrug resistance protein-1 (MRP-1), aquaporin-4 (AQP-4) and cAMP levels were induced by the LiCl treatment, which exhibited more pronounced effects compared with the combination treatment. LiCl was less effective in decreasing the MK and B cell lymphoma-2 (Bcl-2) levels compared with the combination treatment. The most substantial decrease in the p170 levels was identified following the combination treatment, whereas IM induced the second greatest decrease. LiCl alone had no effect on the p170 levels. IM induced the most substantial decrease in the phospho-glycogen synthase kinase 3-beta (p-GSK-3ß)/glycogen synthase kinase 3-beta (GSK-3ß) ratio, and LiCl induced the second most substantial decrease. Both LiCl and the combination treatment induced G2 + M arrest, whereas IM induced G0 + G1 arrest after 72 h of exposure. An apoptotic appearance and autophagic vacuoles were commonly identified in the LiCl, combination and IM groups, respectively. CONCLUSIONS: The combination of IM and LiCl exhibited an antagonist effect, and MK had a role at this antagonism.


Asunto(s)
Antimaníacos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mesilato de Imatinib/farmacología , Cloruro de Litio/farmacología , Acuaporina 4/metabolismo , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , AMP Cíclico/metabolismo , Combinación de Medicamentos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/patología , Glioblastoma/ultraestructura , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Tiempo
17.
Cell Mol Neurobiol ; 35(2): 175-87, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25212824

RESUMEN

Glioblastoma multiforme (GBM) is one of the deadliest human malignancies. A cure for GBM remains elusive, and the overall survival time is less than 1 year. Thus, the development of more efficient therapeutic approaches for the treatment of these patients is required. Induction of tumor cell death by certain phytochemicals derived from medicinal herbs and dietary plants has become a new frontier for cancer therapy research. Although the cancer suppressive effect of Ficus carica (fig) latex (FCL) has been determined in a few cancer types, the effect of this latex on GBM tumors has not been investigated. Therefore, in the current study, the anti-proliferative activity of FCL and the effect of the FCL-temozolomide (TMZ) combination were tested in the T98G, U-138 MG, and U-87 MG GBM cell lines using the WST-1 assay. The mechanism of cell death was analyzed using Annexin-V/FITC and TUNEL assays, and the effect of FCL on invasion was tested using the chick chorioallantoic membrane assay. To determine the effect of FCL on GBM progression, the expression levels of 40 GBM associated miRNAs were analyzed in T98G cells using RT-qPCR. According to the obtained data, FCL causes cell death in GBM cells with different responses to TMZ, and this effect is synergistically increased in combination with TMZ. In addition, the current study is the first to demonstrate the effect of FCL on modulation of let-7d expression, which may be an important underlying mechanism of the anti-invasive effect of this extract.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ficus/química , Glioblastoma/genética , Glioblastoma/patología , Látex/uso terapéutico , MicroARNs/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fraccionamiento Químico , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ , Látex/farmacología , MicroARNs/metabolismo , Modelos Biológicos , Invasividad Neoplásica , Neovascularización Fisiológica/efectos de los fármacos , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Temozolomida
18.
Oral Health Prev Dent ; 13(2): 157-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25197726

RESUMEN

PURPOSE: To evaluate the antibacterial effect of Kenger gum on mutans streptococci (in vivo) and Streptococcus mutans (in vitro) and its cytotoxic effect on the 3T3 fibroblast cell line. MATERIALS AND METHODS: In vitro antibacterial activity of Kenger gum extracts against S.mutans was determined by the disk-diffusion method. The broth dilution method was used to determine the minimum inhibitory concentration (MIC). The cytotoxic effect on 3T3 fibroblast cells at different time intervals was determined using cell culture and viability assays. Clinical studies were then performed on 20 healthy adult subjects, where a sugar-free chewing gum was used as a control. To determine the MS counts, oral rinse samples were taken before chewing as well as 30 and 60 min after 15 min of chewing. Repeated-measures ANOVA was used to compare the bacteria level in the oral rinse samples between the two chewing gums. The Least Significant Difference test was used for adjustment for multiple comparisons. RESULTS: The MIC of the acetone extract of Kenger gum was 30 µg/ml. The acetone extract of Kenger gum possessed moderate antiproliferative properties against the non-tumorigenic cell line 3T3. A statistically significant decrease was observed for both chewing gums at 30 and 60 min. The decrease continued at 60 min after chewing Kenger gum, while the values for control gum tended to approach the baseline after 60 min. CONCLUSION: This preliminary study showed that Kenger gum had particular and prolonged antibacterial activity against S. mutans and salivary mutans streptococci.


Asunto(s)
Antibacterianos/farmacología , Goma de Mascar , Extractos Vegetales/uso terapéutico , Streptococcus mutans/efectos de los fármacos , Células 3T3 , Adolescente , Animales , Asteraceae , Carga Bacteriana/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Goma de Mascar/toxicidad , Femenino , Fibroblastos/efectos de los fármacos , Estudios de Seguimiento , Humanos , Masculino , Ensayo de Materiales , Ratones , Pruebas de Sensibilidad Microbiana , Boca/microbiología , Extractos Vegetales/toxicidad , Adulto Joven
19.
Implant Dent ; 23(6): 641-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25343319

RESUMEN

PURPOSE: Hemostatic agents may be used topically to control hemorrhage, especially in patients with bleeding disorders. The agent used may have a negative effect on the tissue prolonging the healing time. The aim of this study was to compare the effects of 3 different hemostatic agents on fibroblast cells on a rat primary fibroblast cell culture model. MATERIALS AND METHODS: Ankaferd Blood Stopper (ABD) (Ankaferd Pharmaceuticals Cosmetics Production and Marketing Co.), fibrin glue, and tranexamic acid were the agents to be evaluated for their effects on cell proliferation, cell numbers, cell viability, and cell morphology. Also lactate dehydrogenase, basic fibroblast growth factor, and vascular endothelial growth factor C levels were measured. RESULTS: It was found that all of the agents used in the study have negative effects on fibroblasts, with ABD having the lowest values of cell proliferation, cell number, and cell viability. CONCLUSION: The results of this study indicate that ABD, fibrin glue, and tranexamic acid may negatively affect tissue healing.


Asunto(s)
Fibroblastos/efectos de los fármacos , Hemostáticos/farmacología , Extractos Vegetales/farmacología , Animales , Biomarcadores/análisis , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Adhesivo de Tejido de Fibrina/farmacología , Factor 2 de Crecimiento de Fibroblastos/análisis , L-Lactato Deshidrogenasa/análisis , Microscopía Electrónica de Rastreo , Ratas , Ratas Sprague-Dawley , Ácido Tranexámico/farmacología , Factor A de Crecimiento Endotelial Vascular/análisis
20.
Oncol Lett ; 7(6): 2103-2109, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24932297

RESUMEN

Cancer stem cells (CSC) isolated from multiple tumor types differentiate in vivo and in vitro when cultured in serum; however, the factors responsible for their differentiation have not yet been identified. The first aim of the present study was to identify CD133high/CD44high DU145 prostate CSCs and compare their profiles with non-CSCs as bulk counterparts of the population. Subsequently, the two populations continued to be three-dimensional multicellular spheroids. Differentiation was then investigated with stem cell-related genomic characteristics. Polymerase chain reaction array analyses of cell cycle regulation, embryonic and mesenchymal cell lineage-related markers, and telomerase reverse transcriptase (TERT) and Notch signaling were performed. Immunohistochemistry of CD117, Notch1, Jagged1, Delta1, Sox2, c-Myc, Oct4, KLF4, CD90 and SSEA1 were determined in CSC and non-CSC monolayer and spheroid subcultures. Significant gene alterations were observed in the CD133high/CD44high population when cultured as a monolayer and continued as spheroid. In this group, marked gene upregulation was determined in collagen type 9 α1, Islet1 and cyclin D2. Jagged1, Delta-like 3 and Notch1 were respectively upregulated genes in the Notch signaling pathway. According to immunoreactivity, the staining density of Jagged1, Sox2, Oct4 and Klf-4 increased significantly in CSC spheroids. Isolated CSCs alter their cellular characterization over the course of time and exhibit a differentiation profile while maintaining their former surface antigens at a level of transcription or translation. The current study suggested that this differentiation process may be a mechanism responsible for the malignant process and tumor growth.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA