Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Oncotarget ; 8(1): 1438-1448, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27903965

RESUMO

Hyaluronic acid (HA) is abundant in tumor microenvironment and closely associated with invasiveness of glioblastoma (GBM) cells. However, the cellular mechanism underlying HA-rich microenvironment in GBM remains unexplored. Here, we show that tumor-associated mesenchymal stem-like cells (tMSLCs) contribute to abundance of hyaluronic acid (HA) in tumor microenvironment through HA synthase-2 (HAS2) induction, and thereby enhances invasiveness of GBM cells. In an autocrine manner, C5a secreted by tMSLCs activated ERK MAPK for HAS2 induction in tMSLCs. Importantly, HA acted as a signaling ligand of its cognate receptor RHAMM for intracellular signaling activation underlying invasiveness of GBM cells. Taken together, our study suggests that tMSLCs contribute to HA-rich proinvasive ECM microenvironment in GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Células-Tronco Mesenquimais/metabolismo , Idoso , Animais , Neoplasias Encefálicas/patologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Glioblastoma/patologia , Xenoenxertos , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/farmacologia , Ligantes , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transdução de Sinais
11.
Oncotarget ; 7(33): 53430-53442, 2016 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-27462787

RESUMO

Epithelial to mesenchymal transition (EMT) is developmental process associated with cancer metastasis. Here, we found that breast carcinoma cells adopt epithelial-to-mesenchymal transition (EMT) in response to fractionated-radiation. Importantly, we show that Notch signaling is highly activated in fractionally-irradiated tumors as compared to non-irradiated tumors that are accompanied by an EMT. Moreover, we uncovered the mechanism of Notch-driven EMT, in which Notch enhanced EMT through IL-6/JAK/STAT3 signaling axis in mammary tumor cells. Collectively, we present converging evidence from our studies that Notch2 is a critical mediator of radiation-induced EMT and responsible for induced malignant tumor growth.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/efeitos da radiação , Receptor Notch2/metabolismo , Transdução de Sinais/efeitos da radiação , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Radioterapia/efeitos adversos
12.
Oncotarget ; 7(4): 4890-902, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26700818

RESUMO

Poor prognosis of glioblastoma (GBM) is attributable to the propensity of tumor cells to infiltrate into the brain parenchyma. Protein kinase C (PKC) isozymes are highly expressed or aberrantly activated in GBM. However, how this signaling node translates to GBM cell invasiveness remains unknown. Here, we report that among PKC isoforms, PKCδ is strongly associated with infiltration of GBM cells. Notably, PKCδ enhanced Tyr418 phosphorylation of the non-receptor tyrosine kinase SRC, which in turn activated STAT3 and subsequent NOTCH2 signaling, ultimately leading to GBM cell invasiveness. Furthermore, we showed that PKCδ was aberrantly activated in GBM cells by c-MET, a receptor tyrosine kinase hyperactivated in GBM. In agreement, inhibition either component in the c-MET/PKCδ/SRC/STAT3 signaling axis effectively blocked the NOTCH2 signaling and invasiveness of GBM cells. Taken together, our findings shed a light on the signaling mechanisms behind the constitutive activation of PKCδ signaling in GBM.


Assuntos
Biomarcadores Tumorais/metabolismo , Glioblastoma/patologia , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptor Notch2/metabolismo , Idoso , Animais , Apoptose , Biomarcadores Tumorais/genética , Western Blotting , Movimento Celular , Proliferação de Células , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína Quinase C-delta/genética , Proteínas Proto-Oncogênicas c-met/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Sci Rep ; 5: 15809, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26515758

RESUMO

Recently low dose irradiation has gained attention in the field of radiotherapy. For lack of understanding of the molecular consequences of low dose irradiation, there is much doubt concerning its risks on human beings. In this article, we report that low dose irradiation is capable of blocking the oncogenic KRAS-induced malignant transformation. To address this hypothesis, we showed that low dose irradiation, at doses of 0.1 Gray (Gy); predominantly provide defensive response against oncogenic KRAS -induced malignant transformation in human cells through the induction of antioxidants without causing cell death and acts as a critical regulator for the attenuation of reactive oxygen species (ROS). Importantly, we elucidated that knockdown of antioxidants significantly enhanced ROS generation, invasive and migratory properties and abnormal acini formation in KRAS transformed normal as well as cancer cells. Taken together, this study demonstrates that low dose irradiation reduces the KRAS induced malignant cellular transformation through diminution of ROS. This interesting phenomenon illuminates the beneficial effects of low dose irradiation, suggesting one of contributory mechanisms for reducing the oncogene induced carcinogenesis that intensify the potential use of low dose irradiation as a standard regimen.


Assuntos
Transformação Celular Neoplásica/efeitos da radiação , Raios gama , Proteínas ras/genética , Apoptose/efeitos da radiação , Catalase/antagonistas & inibidores , Catalase/genética , Catalase/metabolismo , Linhagem Celular , Movimento Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Transição Epitelial-Mesenquimal , Glutationa Peroxidase/antagonistas & inibidores , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Células MCF-7 , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas ras/metabolismo
14.
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
15.
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
16.
Arch Pharm Res ; 38(3): 408-13, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25620212

RESUMO

Radiotherapy is a standard treatment for many cancers and is frequently used as primary or adjuvant therapy, often in combination with surgery or chemotherapy or both. However, locoregional recurrence or metastatic spread still occurs in a high proportion of patients after radiotherapy. In this regard, emerging evidences suggest that sublethal radiation paradoxically promotes expansion of cancer stem cell population that is highly tumorigenic and is reminiscent of non-neoplasm stem cells. In this review, we discussed recent findings that demonstrate the increase in cancer stem cells after irradiation, and the possible cellular mechanisms with a perspective of tumor microenvironment. A further understating on the mechanistic mechanisms underlying radiation-enhanced malignant phenotypes might increase the efficacy of radiotherapy for cancer treatment.


Assuntos
Neoplasias/radioterapia , Células-Tronco Neoplásicas/metabolismo , Microambiente Tumoral/efeitos da radiação , Terapia Combinada , Humanos , Recidiva Local de Neoplasia , Neoplasias/patologia , Fenótipo , Doses de Radiação , Lesões por Radiação/patologia
17.
Exp Mol Med ; 47: e137, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25633745

RESUMO

Basal-type breast cancers are among the most aggressive and deadly breast cancer subtypes, displaying a high metastatic ability associated with mesenchymal features. However, the molecular mechanisms underlying the maintenance of mesenchymal phenotypes of basal-type breast cancer cells remain obscure. Here, we report that KRAS is a critical regulator for the maintenance of mesenchymal features in basal-type breast cancer cells. KRAS is preferentially activated in basal-type breast cancer cells as compared with luminal type. By loss and gain of KRAS, we found that KRAS is necessary and sufficient for the maintenance of mesenchymal phenotypes and metastatic ability through SLUG expression. Taken together, this study demonstrates that KRAS is a critical regulator for the metastatic behavior associated with mesenchymal features of breast cancer cells, implicating a novel therapeutic target for basal-type breast cancer.


Assuntos
Neoplasias da Mama/genética , Transição Epitelial-Mesenquimal/genética , Proteínas Proto-Oncogênicas/genética , Ativação Transcricional , Proteínas ras/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Fenótipo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/metabolismo
18.
Mol Cell Oncol ; 2(1): e968059, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27308397

RESUMO

The mechanism underlying KRAS (Kirsten rat sarcoma viral oncogene homolog)-driven cellular transformation remains unclear because of the complexity of its downstream effectors. Park et al. recently reported that levels of reactive oxygen species (ROS) are increased by KRAS and are responsible for KRAS-driven malignant transformation, and further identified the signaling cascade involved as KRAS/p38/PDPK1/PKCδ/p47(phox)/NOX1. These findings provide new insight into the molecular mechanisms governing KRAS-driven malignant transformation.

19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA