Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Ann Surg Oncol ; 24(1): 159-166, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27448119

RESUMO

OBJECTIVE: The aim of this study was to analyze clinical and laboratory variables associated with complications after gastrectomy for gastric cancer to predict candidates for successful early discharge. METHODS: Consecutive patients undergoing gastrectomy at Seoul National University Hospital from January through December 2013 were identified from a prospective complications database. Clinicopathologic and postoperative laboratory parameters were analyzed to determine variables associated with complications. An additional validation study was performed from March through May 2014. RESULTS: Overall, complications occurred in 180/855 patients (21.1 %). Age >68 years (odds ratio [OR] 1.64), use of an open approach (OR 1.9), and use of combined resection (OR 1.67) were significant independent risk factors for complications (p < 0.05). The postoperative day (POD) 5 to preoperative white blood cell count (WBC) ratio (risk ratio [RR] 2.01), C-reactive protein (CRP) level on POD 5 (RR 1.1), and maximum body temperature on POD 4 (RR 2.36) independently predicted complications in a multivariate analysis (p < 0.05). After establishing an early discharge profile (EDP) based on these six variables, 152/855 patients (17.8 %) were predicted to have an uncomplicated course. Of these, 8/152 (5.3 %) experienced complications. In a validation study of 217 patients, 43/217 (19.8 %) were candidates for early discharge on POD 5, and 3 (7.0 %) had a false-positive EDP. CONCLUSIONS: Patients younger than 68 years of age who underwent laparoscopic gastrectomy without combined resection might be candidates for early discharge on POD 5 if the POD 5 to preoperative WBC ratio is ≤1.2, POD 5 CRP level is ≤5.38 g/mL, and POD 4 body temperature is ≤37.4 °C.


Assuntos
Gastrectomia , Alta do Paciente , Complicações Pós-Operatórias/epidemiologia , Neoplasias Gástricas/cirurgia , Fatores Etários , Idoso , Feminino , Humanos , Tempo de Internação/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , República da Coreia/epidemiologia , Fatores de Risco , Neoplasias Gástricas/patologia , Resultado do Tratamento
2.
Cancer Sci ; 106(6): 718-725, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25854938

RESUMO

Metastasis of breast cancer is promoted by epithelial-mesenchymal transition (EMT). Emerging evidence suggests that STAT3 is a critical signaling node in EMT and is constitutively activated in many carcinomas, including breast cancer. However, its signaling mechanisms underlying persistent activation of STAT3 associated with EMT remain obscure. Here, we report that PIM2 promotes activation of STAT3 through induction of cytokines. Activation of STAT3 caused an increase in PIM2 expression, implicating a positive feedback loop between PIM2 and STAT3. In agreement, targeting of either PIM2, STAT3 or PIM2-dependent cytokines suppressed EMT-associated migratory and invasive properties through inhibition of ZEB1. Taken together, our findings identify the signaling mechanisms underlying the persistent activation of STAT3 and the oncogenic role of PIM2 in EMT in breast cancer.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fator de Transcrição STAT3/fisiologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Feminino , Proteínas de Homeodomínio/fisiologia , Humanos , Interleucina-1alfa/metabolismo , Interleucina-8/metabolismo , Invasividade Neoplásica , Transdução de Sinais , Fatores de Transcrição/fisiologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco
3.
Cancer Sci ; 106(1): 78-85, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25533622

RESUMO

Despite the fact that ionizing radiation (IR) is widely used as a standard treatment for breast cancer, much evidence suggests that IR paradoxically promotes cancer malignancy. However, the molecular mechanisms underlying radiation-induced cancer progression remain obscure. Here, we report that irradiation activates SRC signaling among SRC family kinase proteins, thereby promoting malignant phenotypes such as invasiveness, expansion of the cancer stem-like cell population, and resistance to anticancer agents in breast cancer cells. Importantly, radiation-activated SRC induced SLUG expression and caused epithelial-mesenchymal cell transition through phosphatidylinositol 3-kinase/protein kinase B and p38 MAPK signaling. In agreement, either inhibition of SRC or downstream signaling of p38 MAPK or protein kinase B effectively attenuated radiation-induced epithelial-mesenchymal cell transition along with an increase in the cancer stem-like cell population. In addition, downregulation of SRC also abolished radiation-acquired resistance of breast cancer cells to anticancer agents such as cisplatin, etoposide, paclitaxel, and IR. Taken together, our findings suggest that combining radiotherapy with targeting of SRC might attenuate the harmful effects of radiation and enhance the efficacy of breast cancer treatment.


Assuntos
Quinases da Família src/metabolismo , Neoplasias da Mama , Movimento Celular/efeitos da radiação , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Ativação Enzimática , Transição Epitelial-Mesenquimal/efeitos da radiação , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Cancer Sci ; 106(1): 94-101, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25456733

RESUMO

Metastasis is a challenging clinical problem and the primary cause of death in breast cancer patients. However, there is no therapeutic agent against metastasis of breast cancer cells. Here we report that phloroglucinol, a natural phlorotannin component of brown algae suppresses metastatic ability of breast cancer cells. Treatment with phloroglucinol effectively inhibited mesenchymal phenotypes of basal type breast cancer cells through downregulation of SLUG without causing a cytotoxic effect. Importantly, phloroglucinol decreased SLUG through inhibition of PI3K/AKT and RAS/RAF-1/ERK signaling. In agreement with in vitro data, phloroglucinol was also effective against in vivo metastasis of breast cancer cells, drastically suppressing their metastatic ability to lungs, and extending the survival time of mice. Collectively, our findings demonstrate a novel anticancer activity of phloroglucinol against metastasis of breast cancer cells, implicating its clinical relevance.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Floroglucinol/farmacologia , Animais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/patologia , Movimento Celular , Feminino , Humanos , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Floroglucinol/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Toxicol Appl Pharmacol ; 286(3): 143-50, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25843036

RESUMO

Poor prognosis of breast cancer patients is closely associated with metastasis and relapse. There is substantial evidence supporting that cancer stem-like cells (CSCs) are primarily responsible for relapse in breast cancer after anticancer treatment. However, there is a lack of suitable drugs that target breast cancer stem-like cells (BCSCs). Here, we report that phloroglucinol (PG), a natural phlorotannin component of brown algae, suppresses sphere formation, anchorage-independent colony formation and in vivo tumorigenicity. In line with these observations, treatment with PG also decreased CD44(+) cancer cell population as well as expression of CSC regulators such as Sox2, CD44, Oct4, Notch2 and ß-catenin. Also, treatment with PG sensitized breast cancer cells to anticancer drugs such as cisplatin, etoposide, and taxol as well as to ionizing radiation. Importantly, PG inhibited KRAS and its downstream PI3K/AKT and RAF-1/ERK signaling pathways that regulate the maintenance of CSCs. Taken together, our findings implicate PG as a good candidate to target BCSCs and to prevent the disease relapse.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Floroglucinol/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Floroglucinol/química , Floroglucinol/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
J Biol Chem ; 287(23): 19516-27, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22511756

RESUMO

The prognosis of breast cancer patients is related to the degree of metastasis. However, the mechanisms by which epithelial tumor cells escape from the primary tumor and colonize at a distant site are not entirely understood. Here, we analyzed expression levels of pituitary tumor-transforming gene-1 (PTTG1), a relatively uncharacterized oncoprotein, in patient-derived breast cancer tissues with corresponding normal breast tissues. We found that PTTG1 is highly expressed in breast cancer patients, compared with normal tissues. Also, PTTG1 expression levels were correlated with the degree of malignancy in breast cancer cell lines; the more migratory and invasive cancer cell lines MDA-MB-231 and BT549 displayed the higher expression levels of PTTG1 than the less migratory and invasive MCF7 and SK-BR3 and normal MCF10A cell lines. By modulating PTTG1 expression levels, we found that PTTG1 enhances the migratory and invasive properties of breast cancer cells by inducing epithelial to mesenchymal transition, as evidenced by altered morphology and epithelial/mesenchymal cell marker expression patterns and up-regulation of the transcription factor Snail. Notably, down-regulation of PTTG1 also suppressed cancer stem cell population in BT549 cells by decreasing self-renewing ability and tumorigenic capacity, accompanying decreasing CD44(high) CD24(low) cells and Sox2 expression. Up-regulation of PTTG1 had the opposite effects, increasing sphere-forming ability and Sox2 expression. Importantly, PTTG1-mediated malignant tumor properties were due, at least in part, to activation of AKT, known to be a key regulator of both EMT and stemness in cancer cells. Collectively, these results suggest that PTTG1 may represent a new therapeutic target for malignant breast cancer.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias da Mama/metabolismo , Movimento Celular , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Antígeno CD24/genética , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo/genética , Ativação Enzimática/genética , Feminino , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Securina , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Cancer Sci ; 104(9): 1172-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23714583

RESUMO

Glioblastoma remains an incurable brain disease due to the prevalence of its recurrence. Considerable evidence suggests that glioma stem-like cells are responsible for glioma relapse after treatment, which commonly involves ionizing radiation. Here, we found that fractionated ionizing radiation (2 Gy/day for 3 days) induced glioma stem-like cell expansion and resistance to anticancer treatment such as cisplatin (50 µM) or taxol (500 nM), or by ionizing radiation (10 Gy) in both glioma cell lines (U87, U373) and patient-derived glioma cells. Of note, concomitant increase of nitric oxide production occurred with the radiation-induced increase of the glioma stem-like cell population through upregulation of inducible nitric oxide synthase (iNOS). In line with this observation, downregulation of iNOS effectively reduced the glioma stem-like cell population and decreased resistance to anticancer treatment. Collectively, our results suggest that targeting iNOS in combination with ionizing radiation might increase the efficacy of radiotherapy for glioma treatment.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Glioblastoma/patologia , Glioblastoma/radioterapia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Óxido Nítrico/biossíntese , Idoso , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/metabolismo , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Radioterapia/efeitos adversos , Regulação para Cima
8.
J Cell Sci ; 124(Pt 18): 3084-94, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21878493

RESUMO

Brain tumors frequently recur or progress as focal masses after treatment with ionizing radiation. However, the mechanisms underlying the repopulation of tumor cells after radiation have remained unclear. In this study, we show that cellular signaling from Abelson murine leukemia viral oncogene homolog (Abl) to protein kinase Cδ (PKCδ) is crucial for fractionated-radiation-induced expansion of glioma-initiating cell populations and acquisition of resistance to anticancer treatments. Treatment of human glioma cells with fractionated radiation increased Abl and PKCδ activity, expanded the CD133-positive (CD133(+)) cell population that possesses tumor-initiating potential and induced expression of glioma stem cell markers and self-renewal-related proteins. Moreover, cells treated with fractionated radiation were resistant to anticancer treatments. Small interfering RNA (siRNA)-mediated knockdown of PKCδ expression blocked fractionated-radiation-induced CD133(+) cell expansion and suppressed expression of glioma stem cell markers and self-renewal-related proteins. It also suppressed resistance of glioma cells to anticancer treatments. Similarly, knockdown of Abl led to a decrease in CD133(+) cell populations and restored chemotherapeutic sensitivity. It also attenuated fractionated-radiation-induced PKCδ activation, suggesting that Abl acts upstream of PKCδ. Collectively, these data indicate that fractionated radiation induces an increase in the glioma-initiating cell population, decreases cellular sensitivity to cancer treatment and implicates activation of Abl-PKCδ signaling in both events. These findings provide insights that might prove pivotal in the context of ionising-radiation-based therapeutic interventions for brain tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Glioma/metabolismo , Glioma/radioterapia , Proteína Quinase C-delta/metabolismo , Antígeno AC133 , Animais , Antígenos CD/biossíntese , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Processos de Crescimento Celular/genética , Processos de Crescimento Celular/efeitos da radiação , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Resistencia a Medicamentos Antineoplásicos/genética , Glioma/tratamento farmacológico , Glioma/patologia , Glicoproteínas/biossíntese , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Células-Tronco Neoplásicas/efeitos da radiação , Proteínas Oncogênicas v-abl/genética , Proteínas Oncogênicas v-abl/metabolismo , Peptídeos , Proteína Quinase C-delta/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Carga Tumoral/genética
9.
Mol Pharmacol ; 82(3): 400-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22648970

RESUMO

Glioma cells with stem cell properties, termed glioma stem-like cells (GSCs), have been linked to tumor formation, maintenance, and progression and are responsible for the failure of chemotherapy and radiotherapy. Because conventional glioma treatments often fail to eliminate GSCs completely, residual surviving GSCs are able to repopulate the tumor. Compounds that target GSCs might be helpful in overcoming resistance to anticancer treatments in human brain tumors. In this study, we showed that 5-bromo-3-(3-hydroxyprop-1-ynyl)-2H-pyran-2-one (BHP), a new 2-pyrone derivative, suppressed the maintenance of the GSC population in both a glioma cell line and patient-derived glioma cells. Treatment of GSCs with BHP effectively inhibited sphere formation and suppressed the CD133(+) cell population. Treatment with BHP also suppressed expression of the stemness-regulating transcription factors Sox2, Notch2, and ß-catenin in sphere-cultured glioma cells. Treatment of GSCs with BHP significantly suppressed two fundamental characteristics of cancer stem cells: self-renewal and tumorigenicity. BHP treatment dramatically inhibited clone-forming ability at the single-cell level and suppressed in vivo tumor formation. BHP markedly inhibited both phosphoinositide 3-kinase/Akt and Ras/Raf-1/extracellular signal-regulated kinase signaling, which suggests that one or both of these pathways are involved in BHP-induced suppression of GSCs. In addition, treatment with BHP effectively sensitized GSCs to chemotherapy and radiotherapy. Taken together, these results indicate that BHP targets GSCs and enhances their sensitivity to anticancer treatments and suggest that BHP treatment may be useful for treating brain tumors by eliminating GSCs.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Pironas/farmacologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Glioma/metabolismo , Glioma/patologia , Glicoproteínas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-raf/metabolismo , Receptor Notch2/antagonistas & inibidores , Receptor Notch2/metabolismo , Fatores de Transcrição SOXB1/antagonistas & inibidores , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo , Proteínas ras/antagonistas & inibidores , Proteínas ras/metabolismo
10.
Anticancer Drugs ; 23(1): 43-50, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21849886

RESUMO

Radiation resistance can be overcome by a combination treatment with chemical modifiers. Here, we showed that treatment with 5-bromo-3-(3-hydroxyprop-1-ynyl)-2H-pyran-2-one (BHP), a new 2-pyrone derivative, in combination with ionizing radiation enhances the sensitivity of human cervical cancer cells to ionizing radiation through overproduction of reactive oxygen species (ROS). The combined treatment with BHP and ionizing radiation caused a decrease in clonogenic survival and an increase in apoptotic cell death in cervical cancer cells. The combined treatment promoted conformational activation of Bax and led to mitochondrial apoptotic cell death. The combination treatment also induced a marked increase in intracellular ROS level. Inhibition of ROS attenuated the radiosensitizing effect of BHP, concurrent with a decrease in Bax activation, a decrease in mitochondrial cell death, and an increase in clonogenic survival. These results indicate that BHP synergistically enhances sensitivity of human cervical cancer cells to ionizing radiation through elevation of intracellular ROS and that ROS-dependent Bax activation is critically involved in the increase in apoptotic cell death induced by the combined treatment with BHP and ionizing radiation.


Assuntos
Pironas/farmacologia , Radiossensibilizantes/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/radioterapia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Terapia Combinada , Feminino , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Piranos , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/patologia , Proteína X Associada a bcl-2/metabolismo
11.
Adv Sci (Weinh) ; 9(2): e2102768, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34813169

RESUMO

Despite aggressive clinical treatment, recurrence of glioblastoma multiforme (GBM) is unavoidable, and the clinical outcome is still poor. A convincing explanation is the phenotypic transition of GBM cells upon aggressive treatment such as radiotherapy. However, the microenvironmental factors contributing to GBM recurrence after treatment remain unexplored. Here, it is shown that radiation-treated GBM cells produce soluble intercellular adhesion molecule-1 (sICAM-1) which stimulates the infiltration of macrophages, consequently enriching the tumor microenvironment with inflammatory macrophages. Acting as a paracrine factor, tumor-derived sICAM-1 induces macrophages to secrete wingless-type MMTV integration site family, member 3A (WNT3A), which promotes a mesenchymal shift of GBM cells. In addition, blockade of either sICAM-1 or WNT3A diminishes the harmful effect of radiation on tumor progression. Collectively, the findings indicate that cellular crosstalk between GBM and macrophage through sICAM-1-WNT3A oncogenic route is involved in the mesenchymal shift of GBM cells after radiation, and suggest that radiotherapy combined with sICAM-1 targeted inhibition would improve the clinical outcome of GBM patients.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos/metabolismo , Mesoderma/metabolismo , Animais , Neoplasias Encefálicas/genética , Modelos Animais de Doenças , Glioblastoma/genética , Humanos , Masculino , Camundongos , Camundongos Nus
12.
Toxicol Appl Pharmacol ; 254(1): 32-40, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21514314

RESUMO

A subpopulation of cancer cells with stem cell properties is responsible for tumor maintenance and progression, and may contribute to resistance to anticancer treatments. Thus, compounds that target cancer stem-like cells could be usefully applied to destroy cancer. In this study, we investigated the effect of Eckol, a phlorotannin compound, on stemness and malignancies in glioma stem-like cells. To determine whether Eckol targets glioma stem-like cells, we examined whether Eckol treatment could change the expression levels of glioma stem-like cell markers and self-renewal-related proteins as well as the sphere forming ability, and the sensitivity to anticancer treatments. Alterations in the malignant properties of sphere-derived cells by Eckol were also investigated by soft-agar colony forming assay, by xenograft assay in nude mice, and by cell invasion assay. Treatment of sphere-forming glioma cells with Eckol effectively decreased the sphere formation as well as the CD133(+) cell population. Eckol treatment suppressed expression of the glioma stem-like cell markers and the self-renewal-related proteins without cell death. Moreover, treatment of glioma stem-like cells with Eckol significantly attenuated anchorage-independent growth on soft agar and tumor formation in xenograft mice. Importantly, Eckol treatment effectively reduced the resistance of glioma stem-like cells to ionizing radiation and temozolomide. Treatment of glioma stem-like cells with Eckol markedly blocked both phosphoinositide 3-kinase-Akt and Ras-Raf-1-Erk signaling pathways. These results indicate that the natural phlorotannin Eckol suppresses stemness and malignancies in glioma stem-like cells, and thereby makes glioma stem-like cells more sensitive to anticancer treatments, providing novel therapeutic strategies targeting specifically cancer stem-like cells.


Assuntos
Antineoplásicos/farmacologia , Dioxinas/farmacologia , Glioma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo
13.
Anticancer Drugs ; 22(8): 763-73, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21642840

RESUMO

A combined treatment with conventional chemotherapies can enhance the effectiveness of chemotherapeutic agents against cancers. Here, we have shown that the naturally occurring triterpenoids synergistically enhance the response of cervical cancer cells to taxol. Of the triterpenoid compounds, pristimerin enhanced the anticancer effect of taxol with the highest efficiency by combination. Pristimerin synergizes with taxol to inhibit clonogenic survival and tumor growth in nude mice, and to enhance cell death in cervical cancer cells. A combined treatment with taxol and pristimerin induced cervical cancer cell death by increasing intracellular reactive oxygen species levels, upregulation of death receptor death receptor 5 (DR5), activation of Bax, and dissipation of mitochondrial membrane potential. Treatment with N-acetyl-L-cysteine, a thiol-containing antioxidant completely blocked combined treatment-induced Bax translocation as well as DR5 upregulation. Moreover, inhibition of Jun N-terminal kinase/c-Jun pathway attenuated cell death by blocking DR5 upregulation and Bax activation. These results indicate that the triterpenoid, pristimerin, synergistically enhances taxol response of cervical cancer cells through DR5 expression and Bax activation. Furthermore, the reactive oxygen species-dependent activation of the Jun N-terminal kinase/c-Jun pathway is required for the DR5 upregulation and Bax activation. The molecular mechanism revealed by this study may aid in the design of future combination cancer therapies against cells with intrinsically reduced sensitivity to taxol.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Morte Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/administração & dosagem , Triterpenos Pentacíclicos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Triterpenos/administração & dosagem , Regulação para Cima/efeitos dos fármacos , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/metabolismo
14.
Mol Cell Neurosci ; 44(4): 318-29, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20417282

RESUMO

Precursors within the subventricular zone (SVZ) exhibit regional variations in the expression of transcription factors important for the regulation of their proliferation and differentiation. In the anterior SVZ (aSVZ) the homeobox transcription factor distalless (Dlx)2 modulates both processes by promoting neural stem cell (NSC) activation as well as neurogenesis. Activated NSCs and transit-amplifying precursors (TAPs) in the aSVZ both express high levels of epidermal growth factor receptor (EGFR(high)) and form clones in response to exogenous EGF. EGF-responsive cells are also present in the hippocampal subependyma (hSVZ). However, it is not clear whether they represent NSCs or TAPs and whether their proliferation and differentiation are regulated as in the aSVZ. Here we have purified EGFR(high) cells from both the aSVZ and hSVZ at different ages. When isolated from perinatal tissue both populations were enriched in multipotent clonogenic precursors, which generated GABAergic neurons. Although they differed in absolute expression levels, activated NSCs and TAPs in both regions displayed similar signatures of transcription factor expression. However, activated NSCs were less frequent in the hSVZ than in the aSVZ. Furthermore, increasing age had a greater inhibitory effect on NSC proliferation in the hSVZ than in the aSVZ. This suggests that NSC activation is differentially regulated in the two regions. Consistent with this hypothesis, we found that in hippocampal precursors Dlx2 promoted neurogenesis but not NSC activation. Thus, most clonogenic EGFR(high) precursors in the hSVZ represent TAPs and NSC proliferation in the aSVZ and hSVZ is regulated by different mechanisms.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Receptores ErbB/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Especificidade de Órgãos , Gravidez , Nicho de Células-Tronco/citologia
15.
Mol Cell Neurosci ; 42(4): 308-14, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19683576

RESUMO

In the postnatal subventricular zone (SVZ) neural stem cells (NSCs) give rise to transit-amplifying precursors (TAPs) expressing high levels of epidermal growth factor receptor (EGFR) that in turn generate neuroblasts. Both TAPs and neuroblasts express distal-less (DLX)2 homeobox transcription factor but the latter proliferate less. Modulation of its expression in vivo has revealed that DLX2 affects both neurogenesis and proliferation in the postnatal SVZ. However, the mechanisms underlying these effects are not clear. To investigate this issue we have here forced the expression of DLX2 in SVZ isolated NSCs growing in defined in vitro conditions. This analysis revealed that DLX2 affects the proliferation of SVZ precursors by regulating two distinct steps of neural lineage progression. Firstly, it promotes the lineage transition from NSCs to TAPs. Secondly it enhances the proliferative response of neuronal progenitors to EGF. Thus DLX2 and EGFR signalling interact at multiple levels to coordinate proliferation in the postnatal SVZ.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Proteínas de Homeodomínio/metabolismo , Neurogênese/fisiologia , Neurônios/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Proteínas de Homeodomínio/genética , Camundongos , Neurônios/citologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/citologia , Células-Tronco/citologia , Fatores de Transcrição/genética
16.
Neuro Oncol ; 22(10): 1452-1462, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32179921

RESUMO

BACKGROUND: Mesenchymal stemlike cells (MSLCs) have been detected in many types of cancer including brain tumors and have received attention as stromal cells in the tumor microenvironment. However, the cellular mechanisms underlying their participation in cancer progression remain largely unexplored. The aim of this study was to determine whether MSLCs have a tumorigenic role in brain tumors. METHODS: To figure out molecular and cellular mechanisms in glioma invasion, we have cultured glioma with MSLCs in a co-culture system. RESULTS: Here, we show that MSLCs in human glioblastoma (GBM) secrete complement component C5a, which is known for its role as a complement factor. MSLC-secreted C5a increases expression of zinc finger E-box-binding homeobox 1 (ZEB1) via activation of p38 mitogen-activated protein kinase (MAPK) in GBM cells, thereby enhancing the invasion of GBM cells into parenchymal brain tissue. CONCLUSION: Our results reveal a mechanism by which MSLCs undergo crosstalk with GBM cells through the C5a/p38 MAPK/ZEB1 signaling loop and act as a booster in GBM progression. KEY POINTS: 1. MSLCs activate p38 MAPK-ZEB1 signaling in GBM cells through C5a in a paracrine manner, thereby boosting the invasiveness of GBM cells in the tumor microenvironment.2. Neutralizing of C5a could be a potential therapeutic target for GBM by inhibition of mesenchymal phenotype.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Células-Tronco Mesenquimais , Linhagem Celular Tumoral , Complemento C5a/genética , Humanos , Invasividade Neoplásica , Microambiente Tumoral , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
17.
Arch Pharm Res ; 42(1): 40-47, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30515725

RESUMO

Cancer is a systemic disease in which neoplastic cells interact with multiple types of non-neoplastic stromal cells as well as non-cellular components. The extracellular matrix (ECM) is a non-cellular component that is aberrantly regulated in many types of tumor microenvironments. Since the ECM generally maintains the tissue structure and provides mechanical forces in the tumor microenvironment, it has been simply assumed to act as a physical barrier for cancer metastasis and have a passive role in cancer progression. However, a substantial body of evidence has suggested that ECM remodeling influences many aspects of cancer cell behaviors and its importance has attracted attention in cancer biology. Abnormal ECM affects cancer progression through several ways such as inducing hypoxia, immune cells interaction by promoting mesenchymal shift and cell transformation. Accordingly, in this review we summarize and discusses the role of the ECM in modulating epithelial cells and surrounding stomatal cell components and considers its prospects in cancer biology.


Assuntos
Progressão da Doença , Matriz Extracelular/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral/fisiologia , Animais , Matriz Extracelular/imunologia , Matriz Extracelular/patologia , Humanos , Imunidade Celular/fisiologia , Invasividade Neoplásica/imunologia , Invasividade Neoplásica/patologia , Neoplasias/imunologia , Neoplasias/patologia
18.
BMB Rep ; 51(4): 182-187, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29301607

RESUMO

In carcinoma, cancer-associated fibroblasts participate in force-mediated extracellular matrix (ECM) remodeling, consequently leading to invasion of cancer cells. Likewise, the ECM remodeling actively occurs in glioblastoma (GBM) and the consequent microenvironmental stiffness is strongly linked to migration behavior of GBM cells. However, in GBM the stromal cells responsible for force-mediated ECM remodeling remain unidentified. We show that tumor-associated mesenchymal stem-like cells (tMSLCs) provide a proinvasive matrix condition in GBM by force-mediated ECM remodeling. Importantly, CCL2-mediated Janus kinase 1 (JAK1) activation increased phosphorylation of myosin light chain 2 in tMSLCs and led to collagen assembly and actomyosin contractility. Collectively, our findings implicate tMSLCs as stromal cells providing force-mediated proinvasive ECM remodeling in the GBM microenvironment, and reminiscent of fibroblasts in carcinoma. [BMB Reports 2018; 51(4): 182-187].


Assuntos
Matriz Extracelular/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patologia , Actomiosina/metabolismo , Idoso , Neoplasias Encefálicas/patologia , Miosinas Cardíacas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Matriz Extracelular/genética , Feminino , Fibroblastos/metabolismo , Humanos , Janus Quinase 1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Cadeias Leves de Miosina/metabolismo , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , Fosforilação , Microambiente Tumoral
19.
Oncogene ; 37(14): 1857-1868, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29348460

RESUMO

Basal type breast cancer is the most aggressive and has mesenchymal features with a high metastatic ability. However, the signaling node that determines the basal type features in breast cancer remains obscure. Here, we report that FYN among SRC family kinases is required for the maintenance of basal type breast cancer subtype. Importantly, FYN enhanced NOTCH2 activation in basal type breast cancer cells through STAT5-mediated upregulation of Jagged-1 and DLL4 NOTCH ligands, thereby contributed to mesenchymal phenotypes. In addition, we found that high levels of FYN persist in basal type breast cancer cells by a positive feedback loop between FYN and STAT5. FYN interacted directly with STAT5 and increased p-STAT5 that further acts as a transcription factor for FYN. Taken together, our findings demonstrate a pivotal role of FYN and its downstream effectors in maintaining the basal type features in breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/genética , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Receptor Notch2/fisiologia , Fator de Transcrição STAT5/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Fenótipo , Proteínas Proto-Oncogênicas c-fyn/genética , Transdução de Sinais/genética
20.
Oncogene ; 37(24): 3317-3328, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29559744

RESUMO

Ionizing radiation is widely used for patient with glioblastoma (GBM). However, the effect of radiation on patient survival is marginal and upon recurrence tumors frequently shift toward mesenchymal subtype adopting invasiveness. Here, we show that ionizing radiation affects biomechanical tension in GBM microenvironment and provides proinvasive extracellular signaling cue, hyaluronic acid (HA)-rich condition. In response to radiation, HA production was increased in GBM cells by HA synthase-2 (HAS2) that was transcriptionally upregulated by NF-ĸB. Notably, NF-ĸB was persistently activated by IL-1α-feedback loop, making HA abundance in tumor microenvironment after radiation. Radiation-induced HA abundance causally has been linked to invasiveness of GBM cells by generating movement track as an extracellular matrix, and by acting as a signaling ligand for CD44 receptor, leading to SRC activation, which is sufficient for mesenchymal shift of GBM cells. Collectively, our findings provide an explanation for the frequent brain tumor relapse after radiotherapy, and potential therapeutic targets to block mesenchymal shift upon relapse.


Assuntos
Neoplasias Encefálicas/radioterapia , Matriz Extracelular/efeitos da radiação , Glioblastoma/radioterapia , Ácido Hialurônico/metabolismo , Microambiente Tumoral/efeitos da radiação , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Receptores de Hialuronatos/metabolismo , Hialuronan Sintases/genética , Estimativa de Kaplan-Meier , Masculino , Camundongos Endogâmicos BALB C , NF-kappa B/genética , NF-kappa B/metabolismo , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA