Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells Tissues Organs ; 211(1): 30-40, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34571511

RESUMO

Marine organisms are among the prominent and abundant sources of 1-O-alkyl-sn-glycerols, including chimyl alcohol (CA), batyl alcohol and selachyl alcohol. These biologically active compounds are precursors in plasmalogen and phospholipid biosynthesis, which are the main irreplaceable components of cell membranes. The results of this study demonstrate that CA promotes the activation of immune processes in the mouse spleen and in the mouse macrophage cell culture RAW 264.7. We studied the effect of CA on the spleen weight, as well as on the proliferation process and expression of interleukin-1ß (IL-1ß) and CD86 - a marker of the proinflammatory M1-type macrophage cell surface in experimental animals. We used the RAW 264.7 macrophage cell culture to assess the CA cytotoxicity, its effect on reactive oxygen species (ROS), nitric oxide (NO) production and lysosomal activity, as well as IL-1ß and CD86 expression in vitro. In vivoand in vitro experiments have shown that CA: (1) enhances cell proliferation without changing the spleen's weight, (2) increases the activity of the proinflammatory marker CD86, (3) increases the level of ROS/NO/lysosome activity, and (4) does not affect the level of IL-1ß. Our study reveals that CA has a complex immunostimulation effect in vitro and in vivo, which opens up prospects for further study of its biological activity.


Assuntos
Macrófagos , Óxido Nítrico , Animais , Éteres de Glicerila , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
Oncol Rep ; 40(5): 2595-2607, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226551

RESUMO

The majority of modern treatment methods for malignant brain tumors are not sufficiently effective, with a median survival time varying between 9 and 14 months. Metastatic and invasive processes are the principal characteristics of malignant tumors. The most important pathogenic mechanism is epithelial­mesenchymal transition (EMT), which causes epithelial cells to become more mobile, and capable of invading the surrounding tissues and migrating to distant organs. Transforming growth factor­ß1 (TGF­ß1) serves a key role in EMT­inducing mechanisms. The current study presented the interaction between hematopoietic stem cells and glioblastoma cells stimulated by TGF­ß1 in vitro. The materials for the study were hematopoietic progenitor cell antigen CD34+ hematopoietic stem cells (HSCs) and U87 glioblastoma cells. Cell culture methods, automated monitoring of cell­cell interactions, confocal laser microscopy, flow cytometry and electron microscopy were used. It was demonstrated that U87 cells have a complex communication system, including adhesive intercellular contacts, areas of interdigitation with dissolution of the cytoplasm, cell fusion, communication microtubes and microvesicles. TGF­ß1 affected glioblastoma cells by modifying the cell shape and intensifying their exocrine function. HSCs migrated to glioblastoma cells, interacted with them and exchanged fluorescent tags. Stimulation of cancer cells with TGF­ß1 weakened the ability of glioblastoma cells to attract HSCs and exchange a fluorescent tag. This process stimulated cancer cell proliferation, which is an indication of the ability of HSCs to 'switch' the proliferation and invasion processes in glioblastoma cells.


Assuntos
Transição Epitelial-Mesenquimal/genética , Glioblastoma/genética , Células-Tronco Hematopoéticas/metabolismo , Fator de Crescimento Transformador beta1/genética , Antígenos CD34/genética , Técnicas de Cultura de Células , Diferenciação Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Células-Tronco Hematopoéticas/patologia , Humanos , Antígenos Comuns de Leucócito/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica
3.
Int J Mol Med ; 42(2): 691-702, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29749540

RESUMO

Glioblastoma multiforme (GBM) is one of the most aggressive brain tumors. GBM represents >50% of primary tumors of the nervous system and ~20% of intracranial neoplasms. Standard treatment involves surgery, radiation and chemotherapy. However, the prognosis of GBM is usually poor, with a median survival of 15 months. Resistance of GBM to treatment can be explained by the presence of cancer stem cells (CSCs) among the GBM cell population. At present, there are no effective therapeutic strategies for the elimination of CSCs. The present review examined the nature of human GBM therapeutic resistance and attempted to systematize and put forward novel approaches for a personalized therapy of GBM that not only destroys tumor tissue, but also regulates cellular signaling and the morphogenetic properties of CSCs. The CSCs are considered to be an informationally accessible living system, and the CSC proteome should be used as a target for therapy directed at suppressing clonal selection mechanisms and CSC generation, destroying CSC hierarchy, and disrupting the interaction of CSCs with their microenvironment and extracellular matrix. These objectives can be achieved through the use of biomedical cellular products.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células-Tronco Neoplásicas/patologia , Medicina de Precisão/métodos , Animais , Tecnologia Biomédica/métodos , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Microambiente Tumoral
4.
Oncol Lett ; 13(2): 738-746, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28356953

RESUMO

Glioblastoma multiforme is an invasive malignant glial brain tumor with a poor prognosis for patients. The primary reasons that lead to the development of treatment resistance are associated with tumor cells infiltrating the brain parenchyma and the specific properties of tumor stem cells. A crucial research area in medical science is the search for effective agents that are able to act on these targets. Fascaplysin alkaloids possess potent antitumor activity. Modern methods for the targeted delivery of drugs reveal extensive possibilities in terms of the clinical use of these compounds. The aim of the present study was to establish effective concentrations of fascaplysin that inhibit the growth and kill the cells of glial tumors, as well as to perform a comparative analysis of fascaplysin's effectiveness in relation to other chemotherapy drugs. C6 glioma cells were utilized as an optimal model of glioblastoma. It was established that fascaplysin at 0.5 µM has a strong cytotoxic effect, which is subsequently replaced by tumor cell death via apoptosis as the length of drug exposure time is increased. Fascaplysin kills glioma cells at a dose higher than 0.5 µM. The efficiency of fascaplysin was observed to significantly exceed that of temozolomide. Therefore, a significant feature of fascaplysin is its ability to inhibit the growth of and kill multipotent tumor cells.

5.
Mol Med Rep ; 14(5): 4511-4520, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748891

RESUMO

Glioblastoma multiforme is an aggressive malignant brain tumor with terminal consequences. A primary reason for its resistance to treatment is associated with cancer stem cells (CSCs), of which there are currently no effective ways to destroy. It remains unclear what cancer cells become a target of stem cell migration, what the role of this process is in oncogenesis and what stem cell lines should be used in developing antitumor technologies. Using modern post­genome technologies, the present study investigated the migration of human stem cells to cancer cells in vitro, the comparative study of cell proteomes of certain stem cells (including CSCs) was conducted and stem cell migration in vivo was examined. Of all glioblastoma cells, CSCs have the stability to attract normal stem cells. Critical differences in cell proteomes allow the consideration of hematopoietic stem cells (HSCs) as an instrument for interaction with glioblastoma CSCs. Following injection into the bloodstream of animals with glioblastoma, the majority of HSCs migrated to the tumor­containing brain hemisphere and penetrated the tumor tissue. HSCs therefore are of potential use in the development of methods to target CSCs.


Assuntos
Transformação Celular Neoplásica/genética , Glioblastoma/genética , Células-Tronco Hematopoéticas/patologia , Neoplasias Experimentais/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Neoplasias Experimentais/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Proteoma/biossíntese , Proteoma/genética , Ratos , Transdução de Sinais/genética
6.
Oncol Lett ; 12(3): 1721-1728, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27602106

RESUMO

The development of antitumor medication based on autologous stem cells is one of the most advanced methods in glioblastoma multiforme (GBM) treatment. However, there are no objective criteria for evaluating the effectiveness of this medication on cancer stem cells (CSCs). One possible criterion could be a change in the number of microglial cells and their specific location in the tumor. The present study aimed to understand the interaction between microglial cells and CSCs in an experimental glioblastoma model. C6 glioma cells were used to create a glioblastoma model, as they have the immunophenotypic characteristics of CSCs. The glioma cells (0.2×106) were stereotactically implanted into the brains of 60 rats. On the 10th, 20th and 30th days after implantation, the animals were 15 of the animals were sacrificed, and the obtained materials were analyzed by morphological and immunohistochemical analysis. Implantation of glioma cells into the rat brains caused rapid development of tumors characterized by invasive growth, angiogenesis and a high rate of proliferation. The maximum concentration of microglia was observed in the tumor nodule between days 10 and 20; a high proliferation rate of cancer cells was also observed in this area. By day 30, necrosis advancement was observed and the maximum number of microglial cells was concentrated in the invasive area; the invasive area also exhibited positive staining for CSC marker antibodies. Microglial cells have a key role in the invasive growth processes of glioblastoma, as demonstrated by the location of CSCs in the areas of microglia maximum concentration. Therefore, the present study indicates that changes in microglia position and corresponding suppression of tumor growth may be objective criteria for evaluating the effectiveness of biomedical treatment against CSCs.

7.
Oncol Rep ; 35(2): 639-48, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26548844

RESUMO

Glioblastoma multiforme (GBM) is one of the most aggressive brain tumors. The majority of modern treatment methods for GBM are not sufficiently effective with a median survival varying from 9 to 14 months. One of the main reasons for the therapeutic resistance of GBM is attributed to cancer stem cells. Pharmaceuticals that can effectively eliminate cancer stem cells do not exist. Experimentally, we have shown that cancer stem cells can be specifically affected to arrest adhesion, proliferation and migration, and other key functions. The main target of this therapy involves membrane intracellular signaling pathways of cancer stem cells that are not subject to neoplastic transformation. An effect on such a complex target requires the development of innovative biotechnological approaches. The research analysis of modern approaches towards creating biomedical drugs for treating cancer stem cells of glioblastoma multiforme is based on advances in the latest cellular and post-genomic technologies. The combination of targeted therapy with regulation of the key functions of cancer stem cells using cell systems with a remodeled proteome is suggested.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Humanos , Terapia de Alvo Molecular/métodos , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Mol Med Rep ; 12(2): 2828-34, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25955107

RESUMO

Glioblastoma multiforme (GM) is an aggressive malignant tumor of the brain. The standard treatment of GM is surgical resection with consequent radio- and chemotherapy with temozolomide. The prognosis is unfavorable, with a survival time of 12-14 months. The phenomenon of targeted migration to the tumor in the brain opens novel possibilities for the treatment of GM. Multipotent mesenchymal stromal cells (MMSCs) are a cell type with anti-carcinogenic properties and can be used to optimize GM therapy. The aim of the present study was to investigate the effects of MMSC transplantation in the chemotherapy of a rat model of C6 glioma. A total of 130 animals were divided into a control group, a temozolomide group, MMSCs group and temozolomide + MMSCs group. The experiment was performed over 70 days, and a combination of molecular biology, surgical and neuroimaging techniques, as well as histological and physiological examinations was used. Tumor size was smallest in the temozolomide (115.76 ± 16.25 mm(3)) and in temozolomide + MMSCs (114.74 ± 5.54 mm(3)) groups, which was significantly smaller than the neoplastic node size in the control group (202.09 ± 39.72 mm(3)) (P<0.05). The animals in the temozolomide + MMSCs group showed significantly higher survival rates in comparison with those in the control and temozolomide groups. The MMSCs migrated from the site of implantation to the neoplastic focus and interacted with glioma cells; however, the mechanism requires further research. In conclusion, MMSC transplantation combined with temozolomide treatment significantly extended the survival of experimental animals in comparison with those treated with temozolomide only.


Assuntos
Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Transplante de Células-Tronco Mesenquimais , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Terapia Combinada , Dacarbazina/uso terapêutico , Modelos Animais de Doenças , Glioblastoma/mortalidade , Glioblastoma/patologia , Masculino , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Wistar , Taxa de Sobrevida , Temozolomida , Transplante Homólogo
9.
Oncol Lett ; 9(4): 1839-1844, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25789053

RESUMO

Multiform glioblastoma is the most common primary, highly invasive, malignant tumor of the central nervous system, with an extremely poor prognosis. The median survival of patients following surgical resection, radiation therapy and chemotherapy does not exceed 12-15 months and thus, novel approaches for the treatment of the disease are required. The phenomenon of the directed migration of stem cells in tumor tissue presents a novel approach for the development of technologies that facilitate the targeted delivery of drugs and other therapeutic agents to the tumor foci. Hematopoietic cluster of differentiation (CD)34+/CD133+ stem cells possess significant reparative potential and are inert with respect to normal neural tissue. The aim of the present study was to investigate the substantiation ability of adult hematopoietic progenitors to the directed migration of glioma cells. A C6 glioma cell line, a culture of hematopoietic CD34+/CD133+ stem cells and primary cultures of rat astrocytes and fibroblasts were used. The cells were co-cultured for five days. The results revealed the formation of cell shaft hematopoietic stem cells on the perimeter of the culture inserts containing the glioma culture. However, this was not observed in the wells with fibroblast and astrocyte cultures. The results indicated that hematopoietic stem cells exhibit a high potential for the directional migration of C6 glioma cells, which allows them to be considered as a promising cell line for the development of novel anticancer biomedical technologies and increases our understanding with regard to previously unclear aspects of glial tumor carcinogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...