Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Clin Transl Med ; 12(8): e997, 2022 08.
Article in English | MEDLINE | ID: mdl-35908277

ABSTRACT

BACKGROUND: The biological function of mesenchymal stem-like cells (MSLCs), a type of stromal cells, in the regulation of the tumour microenvironment is unclear. Here, we investigated the molecular mechanisms underlying extracellular matrix (ECM) remodelling and crosstalk between MSLCs and glioblastomas (GBMs) in tumour progression. METHODS: In vitro and in vivo co-culture systems were used to analyze ECM remodelling and GBM infiltration. In addition, clinical databases, samples from patients with GBM and a xenografted mouse model of GBM were used. RESULTS: Previous studies have shown that the survival of patients with GBM from whom MSLCs could be isolated is substantially shorter than that of patients from whom MSLCs could not be isolated. Therefore, we determined the correlation between changes in ECM-related gene expression in MSLC-isolatable patients with that in MSLC non-isolatable patients using gene set enrichment analysis (GSEA). We found that lysyl oxidase (LOX) and COL1A1 expressions increased in MSLCs via GBM-derived clusters of differentiation 40 ligand (CD40L). Mechanistically, MSLCs are reprogrammed by the CD40L/CD40/NFκB2 signalling axis to build a tumour infiltrative microenvironment involving collagen crosslinking. Importantly, blocking of CD40L by a neutralizing antibody-suppressed LOX expression and ECM remodelling, decreasing GBM infiltration in mouse xenograft models. Clinically, high expression of CD40L, clusters of differentiation 40 (CD40) and LOX correlated with poor survival in patients with glioma. This indicated that GBM-educated MSLCs promote GBM infiltration via ECM remodelling in the tumour microenvironment. CONCLUSION: Our findings provide mechanistic insights into the pro-infiltrative tumour microenvironment produced by GBM-educated MSLCs and highlight a potential therapeutic target that can be used for suppressing GBM infiltration.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CD40 Ligand/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Tumor Microenvironment
2.
Adv Sci (Weinh) ; 9(2): e2102768, 2022 01.
Article in English | MEDLINE | ID: mdl-34813169

ABSTRACT

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.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Macrophages/metabolism , Mesoderm/metabolism , Animals , Brain Neoplasms/genetics , Disease Models, Animal , Glioblastoma/genetics , Humans , Male , Mice , Mice, Nude
3.
Int J Mol Sci ; 22(20)2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34681583

ABSTRACT

Radiation therapy is a current standard-of-care treatment and is used widely for GBM patients. However, radiation therapy still remains a significant barrier to getting a successful outcome due to the therapeutic resistance and tumor recurrence. Understanding the underlying mechanisms of this resistance and recurrence would provide an efficient approach for improving the therapy for GBM treatment. Here, we identified a regulatory mechanism of CD44 which induces infiltration and mesenchymal shift of GBM. Ionizing radiation (IR)-induced K-RAS/ERK signaling activation elevates CD44 expression through downregulation of miR-202 and miR-185 expression. High expression of CD44 promotes SRC activation to induce cancer stemness and EMT features of GBM cells. In this study, we demonstrate that the K-RAS/ERK/CD44 axis is a key mechanism in regulating mesenchymal shift of GBM cells after irradiation. These findings suggest that blocking the K-RAS activation or CD44 expression could provide an efficient way for GBM treatment.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Hyaluronan Receptors/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Movement/radiation effects , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/genetics , Kaplan-Meier Estimate , MicroRNAs/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , RNA, Small Interfering/metabolism
5.
Cancers (Basel) ; 11(2)2019 Feb 10.
Article in English | MEDLINE | ID: mdl-30744156

ABSTRACT

The separation of circulating tumor cells (CTCs) from the peripheral blood is an important issue that has been highlighted because of their high clinical potential. However, techniques that depend solely on tumor-specific surface molecules or just the larger size of CTCs are limited by tumor heterogeneity. Here, we present a slanted weir microfluidic device that utilizes the size and deformability of CTCs to separate them from the unprocessed whole blood. By testing its ability using a highly invasive breast cancer cell line, our device achieved a 97% separation efficiency, while showing an 8-log depletion of erythrocytes and 5.6-log depletion of leukocytes. We also developed an image analysis tool that was able to characterize the various morphologies and differing deformability of the separating cells. From the results, we believe our system possesses a high potential for liquid biopsy, aiding future cancer research.

6.
Micromachines (Basel) ; 9(3)2018 Mar 02.
Article in English | MEDLINE | ID: mdl-30424040

ABSTRACT

Circulating tumor cells (CTCs) are regarded as a strong biomarker which includes clinically valuable information. However, CTCs are very rare and require precise separation and detection for effective clinical applications. Furthermore, downstream analysis has become necessary to identify the distinct sub-population of CTCs that causes metastasis. Here, we report a flow-restricted microfluidic trap array capable of deterministic single-cell capture of CTCs. The extent of flow restriction, correlating with the device geometry, was then optimized using a highly invasive breast cancer cell line (LM2 MDA-MB-231) to achieve 97% capture efficiency with a single-cell capture rate of 99%. Single-cell capture of CTCs from mice with full-blown metastasis was also demonstrated. The single-CTC capturing ability of the flow-restricted trap array not only showed cell enumerating ability but also high prospects for application in future automated downstream analysis.

7.
Oncogene ; 37(43): 5794-5809, 2018 10.
Article in English | MEDLINE | ID: mdl-29973690

ABSTRACT

Understanding the molecular mechanisms that underlie the aggressive behavior and relapse of breast cancer may help in the development of novel therapeutic interventions. CUB-domain-containing protein 1 (CDCP1), a transmembrane adaptor protein, is highly maintained and required in the context of cellular metastatic potential in triple-negative breast cancer (TNBC). For this reason, gene expression levels of CDCP1 have been considered as a prognostic marker in TNBC. However, not rarely, transcript levels of genes do not reflect always the levels of proteins, due to the post-transcriptional regulation. Here we show that miR-17/20a control the FBXL14 E3 ligase, establishing FBXL14 as an upstream regulator of the CDCP1 pathway. FBXL14 acts as an novel interaction partner of CDCP1, and facilitates its ubiquitination and proteasomal degradation with an enhanced capacity to suppress CDCP1 protein stability that eventually prevents CDCP1 target genes involved in breast cancer metastasis. Our findings first time uncovers the regulatory mechanism of CDCP-1 protein stabilization, more predictable criteria than gene expression levels for prognosis of breast cancer patients.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , F-Box Proteins/metabolism , MicroRNAs/genetics , Neoplasm Proteins/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Antigens, CD/genetics , Antigens, Neoplasm , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , F-Box Proteins/genetics , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Prognosis , Transplantation, Heterologous , Triple Negative Breast Neoplasms/mortality , Ubiquitin-Protein Ligases/genetics
8.
Oncogene ; 37(24): 3317-3328, 2018 06.
Article in English | MEDLINE | ID: mdl-29559744

ABSTRACT

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.


Subject(s)
Brain Neoplasms/radiotherapy , Extracellular Matrix/radiation effects , Glioblastoma/radiotherapy , Hyaluronic Acid/metabolism , Tumor Microenvironment/radiation effects , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Hyaluronan Receptors/metabolism , Hyaluronan Synthases/genetics , Kaplan-Meier Estimate , Male , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
9.
Artif Cells Nanomed Biotechnol ; 46(sup2): 87-95, 2018.
Article in English | MEDLINE | ID: mdl-29553850

ABSTRACT

Receptor tyrosine kinase Mer (MerTK) has been shown to be highly expressed in Glioblastoma multiforme (GBM) in comparison to its healthy counterpart and is implicated in brain tumorigenesis. Clarifying the underlying mechanism of MerTK induced invasiveness would result in novel strategies to improve patient's response to chemotherapeutics. In vitro and in vivo assays were performed to examine the functional role of cancer stem sell (CSC) maintenance in MerTK associated invasiveness. In this article, we demonstrate that apart from GBM cells, MerTK is also upregulated in GBM stem-like cells and associated with an increased infiltrative potential of brain tumors in vivo. Silencing of MerTK suppressed the self-renewal of patient-derived GBM stem-like cells. The signaling mechanisms by which MerTK contributes to CSC maintenance have largely been obscure. Molecular analyses revealed that high expression of the signal transducer and activator of transcription 3 (STAT3)- Kirsten rat sarcoma viral oncogene homolog (KRAS) and proto-oncogene tyrosine-protein kinase SRC axis supports MerTK-induced CSC maintenance in GBM spheroids. Furthermore, a short-hairpin RNA-mediated MerTK knockdown effectively blocked invasiveness and N-cadherin expression in mouse xenografts. Collectively, our results uncover a critical function of MerTK in CSC maintenance. Considering the low basal level of MerTK expression in healthy brain cells, evaluation of MerTK as a therapeutic target should advance the research into better therapeutics for GBM.


Subject(s)
Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , c-Mer Tyrosine Kinase/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neoplasm Invasiveness , Prognosis , Proto-Oncogene Mas , c-Mer Tyrosine Kinase/deficiency , c-Mer Tyrosine Kinase/genetics
10.
Oncogene ; 37(14): 1857-1868, 2018 04.
Article in English | MEDLINE | ID: mdl-29348460

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Proto-Oncogene Proteins c-fyn/physiology , Receptor, Notch2/physiology , STAT5 Transcription Factor/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phenotype , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction/genetics
11.
Oncotarget ; 8(47): 83100-83113, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29137327

ABSTRACT

Advanced or progressive cancers share common traits such as altered transcriptional modulation, genetic modification, and abnormal post-translational regulation. These processes influence protein stability and cellular activity. Intercellular adhesion molecule-1 (ICAM-1) is involved in the malignant progression of various human cancers, including breast, liver, renal, and pancreatic cancers, but protein stability has not been deal with in metastatic breast cancer. Additionally, the relevance of the stability maintenance of ICAM-1 protein remains obscure. Here, we identified a novel interaction of E3 ligase FBXO4 that is specifically presented to ICAM-1. To understand how FBXO4 modulates ICAM-1 stability, we investigated ICAM-1-overexpressing or knockdown metastatic/non-metastatic breast cancers. ICAM-1 was found to influence tumor progression and metastasis, whereas FBXO4 regulated aggressive tumorigenic conditions. These results demonstrate that FBXO4 is a major regulator of ICAM-1 stability and that alterations in the stability of ICAM-1 can influence therapeutic outcome in metastatic cancer.

12.
Carbohydr Polym ; 164: 309-316, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28325330

ABSTRACT

The marine mussel-inspired properties of catechol, adhesiveness and cohesiveness, have been applied with pH control to fabricate hollow particles using a silica core and catechol-modified hyaluronic acid (HA-CA) shell for an anticancer drug carrier. The competition between adhesive and cohesive properties of catechol with different pH values leads to various structures, a rough catechol modified HA (HA-CA) shell at pH 5.5, monodisperse spherical silica@HA-CA particles at pH 7.4, and an amorphous HA-CA layer at pH 8.5. The redox transition of catechol with pH is a key factor modulating the behavior of the HA-CA shell on the silica core, which induces strong adhesion of HA-CA to silica at pH 5.5 and structural hardness with cohesive coupling at pH 7.4. In addition, after core removal, the hollow HA-CA particles are followed by loading of anticancer drug, doxorubicin (DOX). DOX loaded HA-CA particles show pH-triggered release behavior and dramatic cytotoxic effect indicating that they are a promising novel anticancer drug carrier.


Subject(s)
Antineoplastic Agents/administration & dosage , Catechols/chemistry , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Cell Line, Tumor , Doxorubicin/administration & dosage , Humans , Hydrogen-Ion Concentration , Silicon Dioxide
13.
Oncotarget ; 8(1): 1438-1448, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27903965

ABSTRACT

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.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Mesenchymal Stem Cells/metabolism , Aged , Animals , Brain Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Female , Glioblastoma/pathology , Heterografts , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/pharmacology , Ligands , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Signal Transduction
14.
Data Brief ; 9: 318-23, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27668278

ABSTRACT

Highly resistant tumor cells are hard to treat at low doses of plasma. Therefore, researchers have gained more attention to development of enhancers for plasma therapy. Some enhancers could improve the efficacy of plasma towards selectivity of cancer cells damage. In this dataset, we report the application of low doses of PEG-coated gold nanoparticles with addition of plasma treatment. This data consists of the effect of PEG-coated GNP and cold plasma on two solid tumor cell lines T98G glioblastoma and A549 lung adenocarcinoma. Cell proliferation, frequency of cancer stem cell population studies by this co-treatment was reported. Finally, we included in this dataset the effect of co-treatment in vivo, using tumor xenograft nude mice models. The data supplied in this article supports the accompanying publication "Low doses of PEG-coated gold nanoparticles sensitize solid tumors to cold plasma by blocking the PI3K/AKT-driven signaling axis to suppress cellular transformation by inhibiting growth and EMT" (N. K. Kaushik, N. Kaushik, K. C. Yoo, N Uddin, J. S. Kim, S. J. Lee et al., 2016) [1].

15.
Oncotarget ; 7(33): 53430-53442, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27462787

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/radiation effects , Receptor, Notch2/metabolism , Signal Transduction/radiation effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Radiotherapy/adverse effects
16.
Biomaterials ; 87: 118-130, 2016 May.
Article in English | MEDLINE | ID: mdl-26921841

ABSTRACT

Metastasis, the primary cause of tumor cell transformation, is often activated during cancer invasion and progression and is associated with poor therapeutic outcomes. The effects of combined treatments that included PEG-coated gold nanoparticles (GNP) and cold plasma on epithelial-mesenchymal transition (EMT) and the maintenance of cancer stem cells (CSC) have not been described so far. Here, we report that co-treatment with GNP and cold plasma inhibited proliferation in cancer cells by abolishing the activation of the PI3K/AKT signaling axis. In addition, co-treatment reversed EMT in solid tumor cells by reducing the secretion of a number of proteins, resulting in the upregulation of epithelial markers such as E-cadherin along with down-regulation of N-Cadherin, Slug and Zeb-1. The inhibition of the PI3K/AKT pathway and the reversal of EMT by co-treatment prevented tumor cells growth in solid tumors. Furthermore, we show that GNP and plasma also suppresses tumor growth by decreasing mesenchymal markers in tumor xenograft mice models. Importantly, co-treatment resulted in a substantial decrease in sphere formation and the self-renewal capacity of glioma-like stem cells. Together, these results indicate a direct link between a decrease of EMT and an increase in cell death in solid tumors following co-treatment with cold plasma and GNP.


Subject(s)
Brain Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Glioma/drug therapy , Gold/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Plasma Gases/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glioma/metabolism , Glioma/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects
17.
Oncotarget ; 7(4): 4890-902, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26700818

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Glioblastoma/pathology , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, Notch2/metabolism , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Cell Movement , Cell Proliferation , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase C-delta/genetics , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Notch2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Sci Rep ; 5: 15809, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26515758

ABSTRACT

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.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Gamma Rays , ras Proteins/genetics , Apoptosis/radiation effects , Catalase/antagonists & inhibitors , Catalase/genetics , Catalase/metabolism , Cell Line , Cell Movement/radiation effects , Dose-Response Relationship, Radiation , Epithelial-Mesenchymal Transition , Glutathione Peroxidase/antagonists & inhibitors , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , MCF-7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , ras Proteins/metabolism
19.
Cancer Sci ; 106(6): 718-725, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25854938

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , STAT3 Transcription Factor/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Female , Homeodomain Proteins/physiology , Humans , Interleukin-1alpha/metabolism , Interleukin-8/metabolism , Neoplasm Invasiveness , Signal Transduction , Transcription Factors/physiology , Zinc Finger E-box-Binding Homeobox 1
20.
Exp Mol Med ; 47: e137, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25633745

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Proto-Oncogene Proteins/genetics , Transcriptional Activation , ras Proteins/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...