Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(29): eadp1439, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39018413

ABSTRACT

Spatiotemporally controllable droplet manipulation is vital across numerous applications, particularly in miniature droplet robots known for their exceptional deformability. Despite notable advancements, current droplet control methods are predominantly limited to two-dimensional (2D) deformation and motion of an individual droplet, with minimal exploration of 3D manipulation and collective droplet behaviors. Here, we introduce a bimodal actuation strategy, merging magnetic and optical fields, for remote and programmable 3D guidance of individual ferrofluidic droplets and droplet collectives. The magnetic field induces a magnetic dipole force, prompting the formation of droplet collectives. Simultaneously, the optical field triggers isothermal changes in interfacial tension through Marangoni flows, enhancing buoyancy and facilitating 3D movements of individual and collective droplets. Moreover, these droplets can function autonomously as soft robots, capable of transporting objects. Alternatively, when combined with a hydrogel shell, they assemble into jellyfish-like robots, driven by sunlight. These findings present an efficient strategy for droplet manipulation, broadening the capabilities of droplet-based robotics.

2.
J Radiat Res ; 63(6): 817-827, 2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36253116

ABSTRACT

The primary motivation of this investigative study is trying to find an alternative treatment that can be used to slow down or treat glioblastoma due to the witnessed toxic side effects of the current drugs coupled with limited effectiveness in overall treatment. Consequently, a Chinese plant extract emodin proves to play a critical role in this investigative study since results from the Western blot and the other accompanying assays for anti-cancer effects indicate that it cannot work a lot to suppress cell migration and possible invasion, but rather emodin can be combined with radiation to give desired outcomes. Our result shows that the kind of radiation which acts well with emodin is neutron radiation rather than gamma radiation. Emodin significantly enhanced the radiosensitivity of LN18 and LN428 cells to γ-rays through MTT assay and cell counting. Accordingly, exposure to neutron radiation in the presence of emodin induced apoptotic cell death and autophagic cell death to a significantly higher extent, and suppressed cell migration and invasiveness more robustly. These effects are presumably due to the ability of emodin to amplify the effective dose from neutron radiation more efficiently. Thus, the study below is one such trial towards new interventional discovery and development in relation to glioblastoma treatment.


Subject(s)
Emodin , Emodin/pharmacology , Emodin/therapeutic use
3.
Pharmacol Res Perspect ; 10(4): e00989, 2022 08.
Article in English | MEDLINE | ID: mdl-35904494

ABSTRACT

Drug repositioning is an alternative process for drug development in cancer. Specifically, it is a strategy for the discovery of new antitumor drugs by screening previously approved clinical drugs. On the basis of this strategy, aripiprazole, an antipsychotic drug, was found to have anticancer activity. In this study, we investigated the radiosensitizing effects of aripiprazole on head and neck cancer cells at sublethal doses of ionizing radiation (IR) in vitro and in vivo. Treatment with aripiprazole suppressed the growth of head and neck cancer cells in a concentration-dependent manner, as evidenced by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Intriguingly, aripiprazole significantly enhanced the sensitivity of these cells to the IC50 dose of IR. The combination of aripiprazole with IR synergistically increased annexin and propidium iodide double-positive and terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cell populations, and induced cleaved poly(ADP-ribose) polymerase and caspase-3 expression, indicating the induction of apoptosis in these cells. Aripiprazole and IR-induced apoptosis were accompanied by an increase in reactive oxygen species and was almost completely suppressed by the addition of the antioxidant, N-acetylcysteine. Finally, aripiprazole greatly sensitized xenograft tumors to IR at doses that did not affect tumor growth. Taken together, these results suggest that aripiprazole could be considered a potent radiosensitizer for head and neck cancer.


Subject(s)
Head and Neck Neoplasms , Aripiprazole/pharmacology , Aripiprazole/therapeutic use , Head and Neck Neoplasms/drug therapy , Humans , In Situ Nick-End Labeling , Radiation, Ionizing , Reactive Oxygen Species/metabolism
4.
J Radiat Res ; 63(3): 342-353, 2022 May 18.
Article in English | MEDLINE | ID: mdl-35446963

ABSTRACT

Glioblastoma is a deadly cancer tumor in the brain and has a survival rate of about 15 months. Despite the high mortality rate, temozolomide has proven to increase the survival rate of patients when combined with radiotherapy. However, its effects may be limited because some patients develop therapeutic resistance. Curcumin has proven to be a cancer treatment due to its broad anticancer spectrum, high efficiency and low toxic level. Additionally, curcumin significantly enhanced radiation efficacy under high and low Linear Energy Transfer (LET) radiation conditions in vitro. In combination with radiation, curcumin increased the cell population in the sub-G1 phase and the reactive oxygen species (ROS) level, ultimately increasing GBM cellular apoptosis. The radiosensitizing effects of curcumin are much higher in neutron (high LET)-irradiated cell lines than in γ (low LET)-irradiated cell lines. Curcumin plus neutron combination significantly inhibited cell invasion compared with that of single treatment or curcumin combined γ-ray treatment. Curcumin enhances the radiosensitivity of Glioblastoma (GBM), suggesting it may have clinical utility in combination cancer treatment with neutron high-LET radiation.


Subject(s)
Curcumin , Glioblastoma , Apoptosis , Cell Line, Tumor , Curcumin/pharmacology , Glioblastoma/pathology , Humans , Linear Energy Transfer , Radiation Tolerance
5.
Transl Oncol ; 15(1): 101255, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34742152

ABSTRACT

The resistance of highly aggressive glioblastoma multiforme (GBM) to chemotherapy is a major clinical problem resulting in a poor prognosis. GBM contains a rare population of self-renewing cancer stem cells (CSCs) that proliferate, spurring the growth of new tumors, and evade chemotherapy. In cancer, major vault protein (MVP) is thought to contribute to drug resistance. However, the role of MVP as CSCs marker remains unknown and whether MVP could sensitize GBM cells to Temozolomide (TMZ) also is unclear. We found that sensitivity to TMZ was suppressed by significantly increasing the MVP expression in GBM cells with TMZ resistance. Also, MVP was associated with the expression of other multidrug-resistant proteins in tumorsphere of TMZ-resistant GBM cell, and was highly co-expressed with CSC markers in tumorsphere culture. On the other hands, knockdown of MVP resulted in reduced sphere formation and invasive capacity. Moreover, high expression of MVP was associated with tumor malignancy and survival rate in glioblastoma patients. Our study describes that MVP is a potentially novel maker for glioblastoma stem cells and may be useful as a target for preventing TMZ resistance in GBM patients.

6.
Sci Rep ; 11(1): 18230, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521931

ABSTRACT

During radiotherapy, microenvironments neighboring the tumor are also exposed to gamma irradiation; this results in unexpected side effects. Blood vessels can serve as microenvironments for tumors and they play an important role in providing nutrients to tumors. This is mostly related to tumor progression, metastasis, and relapse after therapy. Many studies have been performed to obtain a better understanding of tumor vasculature after radiotherapy with in vitro models. However, compared to 3-D models, 2-D in vitro endothelial monolayers cannot physiologically reflect in vivo blood vessels. We previously remodeled the extracellular matrix (ECM) hydrogel that enhanced the tight barrier formation of 3-D blood vessels and the vascular endothelial growth factor (VEGF) gradient induced angiogenesis in a microfluidic device. In this study, the blood vessel model is further introduced to understand how gamma irradiation affects the endothelial monolayer. After the gamma irradiation exposure, we observed a collapsed endothelial barrier and a reduced angiogenic potential. Changes in the cell behaviors of the tip and stalk cells were also detected in the angiogenesis model after irradiation, which is difficult to observe in 2-D monolayer models. Therefore, the 3-D in vitro blood vessel model can be used to understand radiation-induced endothelial injuries.


Subject(s)
Endothelial Cells/radiation effects , Gamma Rays , Neovascularization, Pathologic/metabolism , Tissue Engineering/methods , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Microfluidics/methods , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Brain ; 144(2): 636-654, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33479772

ABSTRACT

As the clinical failure of glioblastoma treatment is attributed by multiple components, including myelin-associated infiltration, assessment of the molecular mechanisms underlying such process and identification of the infiltrating cells have been the primary objectives in glioblastoma research. Here, we adopted radiogenomic analysis to screen for functionally relevant genes that orchestrate the process of glioma cell infiltration through myelin and promote glioblastoma aggressiveness. The receptor of the Nogo ligand (NgR1) was selected as the top candidate through Differentially Expressed Genes (DEG) and Gene Ontology (GO) enrichment analysis. Gain and loss of function studies on NgR1 elucidated its underlying molecular importance in suppressing myelin-associated infiltration in vitro and in vivo. The migratory ability of glioblastoma cells on myelin is reversibly modulated by NgR1 during differentiation and dedifferentiation process through deubiquitinating activity of USP1, which inhibits the degradation of ID1 to downregulate NgR1 expression. Furthermore, pimozide, a well-known antipsychotic drug, upregulates NgR1 by post-translational targeting of USP1, which sensitizes glioma stem cells to myelin inhibition and suppresses myelin-associated infiltration in vivo. In primary human glioblastoma, downregulation of NgR1 expression is associated with highly infiltrative characteristics and poor survival. Together, our findings reveal that loss of NgR1 drives myelin-associated infiltration of glioblastoma and suggest that novel therapeutic strategies aimed at reactivating expression of NgR1 will improve the clinical outcome of glioblastoma patients.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Myelin Sheath/metabolism , Nogo Receptor 1/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Proteins/metabolism , Mice, Inbred BALB C , Myelin Sheath/pathology , Ubiquitin-Specific Proteases/metabolism
9.
Cell Death Dis ; 12(1): 48, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33414415

ABSTRACT

Lung cancer is one of the most common reasons for cancer-induced mortality across the globe, despite major advancements in the treatment strategies including radiotherapy and chemotherapy. Existing reports suggest that CXCR4 is frequently expressed by malignant tumor and is imperative for vascularization, tumor growth, cell migration, and metastasis pertaining to poor prognosis. In this study, we infer that CXCR4 confers resistance to ionizing radiation (IR) in nonsmall cell lung cancer (NSCLC) cells. Further, on the basis of colony forming ability, one finds that drug-resistant A549/GR cells with improved CXCR4 expression exhibited more resistance to IR than A549 cells evidenced along with a reduction in the formation of γ-H2AX foci after IR. Transfection of shRNA against CXCR4 or treatment of pharmacological inhibitor (AMD3100) both led to sensitization of A549/GR cells towards IR. Conversely, the overexpression of CXCR4 in A549 and H460 cell lines was found to improve clonogenic survival, and reduce the formation of γ-H2AX foci after IR. CXCR4 expression was further correlated with STAT3 activation, and suppression of STAT3 activity with siSTAT3 or a specific inhibitor (WP1066) significantly stymied the colony-forming ability and increased γ-H2AX foci formation in A549/GR cells, indicating that CXCR4-mediated STAT3 signaling plays an important role for IR resistance in NSCLC cells. Finally, CXCR4/STAT3 signaling was mediated with the upregulation of Slug and downregulation of the same with siRNA, which heightened IR sensitivity in NSCLC cells. Our data collectively suggests that CXCR4/STAT3/Slug axis is paramount for IR resistance of NSCLC cells, and can be regarded as a therapeutic target to enhance the IR sensitivity of this devastating cancer.


Subject(s)
Receptors, CXCR4/metabolism , STAT3 Transcription Factor/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Mice , Prognosis , Transfection
10.
Oncogene ; 40(8): 1490-1502, 2021 02.
Article in English | MEDLINE | ID: mdl-33452454

ABSTRACT

Glioblastoma multiforme (GBM) or glioblastoma is the most deadly malignant brain tumor in adults. GBM is difficult to treat mainly due to the presence of glioblastoma stem cells (GSCs). Epidermal growth factor receptor variant III (EGFRvIII) has been linked to stemness and malignancy of GSCs; however, the regulatory mechanism of EGFRvIII is largely unknown. Here, we demonstrated that Anoctamin-1 (ANO1), a Ca2+-activated Cl- channel, interacts with EGFRvIII, increases its protein stability, and supports the maintenance of stemness and tumor progression in GSCs. Specifically, shRNA-mediated knockdown and pharmacological inhibition of ANO1 suppressed the self-renewal, invasion activities, and expression of EGFRvIII and related stem cell factors, including NOTCH1, nestin, and SOX2 in GSCs. Conversely, ANO1 overexpression enhanced the above phenomena. Mechanistically, ANO1 protected EGFRvIII from proteasomal degradation by directly binding to it. ANO1 knockdown significantly increased survival in mice and strongly suppressed local invasion of GSCs in an in vivo intracranial mouse model. Collectively, these results suggest that ANO1 plays a crucial role in the maintenance of stemness and invasiveness of GSCs by regulating the expression of EGFRvIII and related signaling molecules, and can be considered a promising therapeutic target for GBM treatment.


Subject(s)
Anoctamin-1/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Animals , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/pathology , Nestin/genetics , Protein Stability , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , SOXB1 Transcription Factors/genetics
11.
Oncogene ; 40(3): 508-521, 2021 01.
Article in English | MEDLINE | ID: mdl-33188296

ABSTRACT

PARK7 is involved in many key cellular processes, including cell proliferation, transcriptional regulation, cellular differentiation, oxidative stress protection, and mitochondrial function maintenance. Deregulation of PARK7 has been implicated in the pathogenesis of various human diseases, including cancer. Here, we aimed to clarify the effect of PARK7 on stemness and radioresistance of glioblastoma stem cells (GSCs). Serum differentiation and magnetic cell sorting of GSCs revealed that PARK7 was preferentially expressed in GSCs rather than differentiated GSCs. Immunohistochemical staining showed enhanced expression of PARK7 in glioma tissues compared to that in normal brain tissues. shRNA-mediated knockdown of PARK7 inhibited the self-renewal activity of GSCs in vitro, as evidenced by the results of neurosphere formation, limiting dilution, and soft-agar clonogenic assays. In addition, PARK7 knockdown suppressed GSC invasion and enhanced GSC sensitivity to ionizing radiation (IR). PARK7 knockdown suppressed expression of GSC signatures including nestin, epidermal growth factor receptor variant III (EGFRvIII), SOX2, NOTCH1, and OCT4. Contrarily, overexpression of PARK7 in CD133- non-GSCs increased self-renewal activities, migration, and IR resistance, and rescued the reduction of GSC factors under shPARK7-transfected and serum-differentiation conditions. Intriguingly, PARK7 acted as a co-chaperone of HSP90 by binding to it, protecting EGFRvIII from proteasomal degradation. Knockdown of PARK7 increased the production of reactive oxygen species, inducing partial apoptosis and enhancing IR sensitivity in GSCs. Finally, PARK7 knockdown increased mouse survival and IR sensitivity in vivo. Based on these data, we propose that PARK7 plays a pivotal role in the maintenance of stemness and therapeutic resistance in GSCs.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/enzymology , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/enzymology , Protein Deglycase DJ-1/metabolism , Cell Line, Tumor , ErbB Receptors/genetics , Glioblastoma/pathology , Humans , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Protein Deglycase DJ-1/genetics
13.
Cells ; 9(5)2020 04 26.
Article in English | MEDLINE | ID: mdl-32357567

ABSTRACT

ANO1, a Ca2+-activated chloride channel, is highly expressed in glioblastoma cells and its surface expression is involved in their migration and invasion. However, the regulation of ANO1 surface expression in glioblastoma cells is largely unknown. In this study, we found that Ca2+/Calmodulin-dependent protein kinase II (CaMKII) ß specifically enhances the surface expression and channel activity of ANO1 in U251 glioblastoma cells. When KN-93, a CaMKII inhibitor, was used to treat U251 cells, the surface expression and channel activity of ANO1 were significantly reduced. Only CaMKIIß, among the four CaMKII isoforms, increased the surface expression and channel activity of ANO1 in a heterologous expression system. Additionally, gene silencing of CaMKIIß suppressed the surface expression and channel activity of ANO1 in U251 cells. Moreover, gene silencing of CaMKIIß or ANO1 prominently reduced the migration and invasion of U251 and U87 MG glioblastoma cells. We thus conclude that CaMKIIß plays a specific role in the surface expression of ANO1 and in the ANO1-mediated tumorigenic properties of glioblastoma cells, such as migration and invasion.


Subject(s)
Anoctamin-1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glioblastoma/metabolism , Neoplasm Proteins/metabolism , Anoctamin-1/genetics , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Humans , Neoplasm Proteins/genetics
14.
Exp Mol Med ; 52(4): 629-642, 2020 04.
Article in English | MEDLINE | ID: mdl-32280134

ABSTRACT

Glioblastomas (GBMs) are characterized by four subtypes, proneural (PN), neural, classical, and mesenchymal (MES) GBMs, and they all have distinct activated signaling pathways. Among the subtypes, PN and MES GBMs show mutually exclusive genetic signatures, and the MES phenotype is, in general, believed to be associated with more aggressive features of GBM: tumor recurrence and drug resistance. Therefore, targeting MES GBMs would improve the overall prognosis of patients with fatal tumors. In this study, we propose peroxisome proliferator-activated receptor gamma (PPARγ) as a potential diagnostic and prognostic biomarker as well as therapeutic target for MES GBM; we used multiple approaches to assess PPARγ, including biostatistics analysis and assessment of preclinical studies. First, we found that PPARγ was exclusively expressed in MES glioblastoma stem cells (GSCs), and ligand activation of endogenous PPARγ suppressed cell growth and stemness in MES GSCs. Further in vivo studies involving orthotopic and heterotopic xenograft mouse models confirmed the therapeutic efficacy of targeting PPARγ; compared to control mice, those that received ligand treatment exhibited longer survival as well as decreased tumor burden. Mechanistically, PPARγ activation suppressed proneural-mesenchymal transition (PMT) by inhibiting the STAT3 signaling pathway. Biostatistical analysis using The Cancer Genomics Atlas (TCGA, n = 206) and REMBRANDT (n = 329) revealed that PPARγ upregulation is linked to poor overall survival and disease-free survival of GBM patients. Analysis was performed on prospective (n = 2) and retrospective (n = 6) GBM patient tissues, and we finally confirmed that PPARγ expression was distinctly upregulated in MES GBM. Collectively, this study provides insight into PPARγ as a potential therapeutic target for patients with MES GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Glioblastoma/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Mice , PPAR gamma/genetics , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Transcriptome , Xenograft Model Antitumor Assays
15.
Int J Oncol ; 56(6): 1540-1550, 2020 06.
Article in English | MEDLINE | ID: mdl-32236607

ABSTRACT

The epidermal growth factor receptor (EGFR)­tyrosine kinase inhibitor (TKI), gefitinib, is an effective therapeutic drug used in the treatment of non­small cell lung cancers (NSCLCs) harboring EGFR mutations. However, acquired resistance significantly limits the efficacy of EGFR­TKIs and consequently, the current chemotherapeutic strategies for NSCLCs. It is, therefore, necessary to overcome this resistance. In the present study, the anticancer potential of natural extracts of Coptis chinensis (ECC) against gefitinib­resistant (GR) NSCLC cells were investigated in vitro and in vivo. ECC inhibited the viability, migration and invasion, and effectively induced the apoptosis of GR cells. These effects were associated with the suppression of EGFR/AKT signaling and the expression of anti­apoptotic proteins, Mcl­1 and Bcl­2, which were overexpressed in GR NSCLC cells. Combination treatment with ECC and gefitinib enhanced the sensitivity of GR cells to gefitinib in vitro, but not in vivo. However, ECC increased the survival of individual zebrafish without affecting the anticancer effect to cancer cells in vivo, which indicated a specific cytotoxic effect of ECC on cancer cells, but not on normal cells; this is an important property for the development of novel anticancer drugs. On the whole, the findings of the present study indicate the potential of ECC for use in the treatment of NSCLC, particularly in combination with EGFR­TKI therapy, in EGFR­TKI­resistant cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Coptis/chemistry , Drug Resistance, Neoplasm/drug effects , Gefitinib/administration & dosage , Lung Neoplasms/drug therapy , Phytochemicals/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drug Synergism , Gefitinib/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Humans , Lung Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Phytochemicals/chemistry , Phytochemicals/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Xenograft Model Antitumor Assays , Zebrafish
16.
Neuro Oncol ; 22(10): 1452-1462, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32179921

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Mesenchymal Stem Cells , Cell Line, Tumor , Complement C5a/genetics , Humans , Neoplasm Invasiveness , Tumor Microenvironment , Zinc Finger E-box-Binding Homeobox 1/genetics , p38 Mitogen-Activated Protein Kinases/genetics
17.
Front Oncol ; 9: 1259, 2019.
Article in English | MEDLINE | ID: mdl-31803626

ABSTRACT

Glioblastoma (GBM) is a largely fatal and highly angiogenic malignancy with a median patient survival of just over 1 year with radiotherapy (RT). The effects of RT on GBM remain unclear, although increasing evidence suggests that RT-induced alterations in the brain microenvironment affect the recurrence and aggressiveness of GBM. Glioma stem cells (GSCs) in GBM are resistant to conventional therapies, including RT. This study aimed to investigate the effect of radiation on tumor growth and the GSC microenvironment in a mouse model of glioma. To evaluate the growth-inhibitory effects of ionizing radiation on GSCs, tumor volume was measured via anatomical magnetic resonance imaging (MRI) after the intracranial injection of 1 × 104 human patient-derived GSCs (83NS cells), which exhibit marked radioresistance. When a tumor mass of ~5 mm3 was detected in each animal, 10 Gy of cranial irradiation was administered. Tumor progression was observed in the orthotopic xenografted GSC tumor (primary tumor) from a detectable tumor mass (5 mm3) to a lethal tumor mass (78 mm3) in ~7 d in the non-irradiated group. In the RT group, tumor growth was halted for almost 2 weeks after administering 10 Gy cranial irradiation, with tumor growth resuming thereafter and eventually approaching a lethal mass (56 mm3) 21 d after radiation. Radiation therapy yielded good therapeutic effects, with a 2-fold increase in GSC glioma survival; however, tumor relapse after RT resulted in higher mortality for the mice with a smaller tumor volume (p = 0.029) than the non-irradiated tumor-bearing mice. Moreover, tumor regrowth after IR resulted in different phenotypes associated with glioma aggressiveness compared with the non-irradiated mice; the apparent diffusion coefficient by diffusion MRI decreased significantly (p < 0.05, 0 Gy vs. 10 Gy) alongside decreased angiogenesis, abnormal vascular dilatation, and upregulated CD34, VWF, AQP1, and AQP4 expression in the tumor. These findings demonstrate that radiation affects GSCs in GBM, potentially resulting in therapeutic resistance by changing the tumor microenvironment. Thus, the results of this study suggest potential therapeutic targets for overcoming the resistance of GBMs to RT.

18.
Exp Mol Med ; 51(10): 1-15, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649250

ABSTRACT

Nogo receptor (NgR) has been shown to inhibit the migration and invasion of human glioma cells. However, little is known regarding the regulatory mechanisms of NgR in glioblastoma multiforme (GBM). In this study, we propose a novel mechanism that regulates the maturation process of NgR through an interaction with vimentin. The inhibition of TGFß1 activity by LY2109761 attenuated the migration/invasion of GBM cells by upregulating cell-surface NgR. Conversely, the treatment of GBM cells with TGFß1 suppressed NgR maturation. We showed that NgR and vimentin interact, which could be a possible mechanism for the suppression of NgR maturation. The knockdown of vimentin suppressed the migration/invasion of GBM cells through the increased maturation of NgR. Finally, TCGA (The Cancer Genome Atlas) analysis also supported the association of NgR and vimentin. The maturation of NgR is regulated by the interaction of vimentin and NgR, which attenuates the invasive activity of GBM, and might be a potential therapeutic target for brain cancer.


Subject(s)
Extracellular Matrix Proteins/genetics , Glioblastoma/genetics , Nogo Receptor 1/genetics , Transforming Growth Factor beta/genetics , Vimentin/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/pathology , Humans , Neoplasm Invasiveness/genetics
19.
Sci Rep ; 9(1): 14040, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31575874

ABSTRACT

Transparent and conducting flexible electrodes have been successfully developed over the last few decades due to their potential applications in optoelectronics. However, recent developments in smart electronics, such as a direct human-machine interface, health-monitoring devices, motion-tracking sensors, and artificially electronic skin also require materials with multifunctional properties such as transparency, flexibility and good portability. In such devices, there remains room to develop transparent and flexible devices such as pressure sensors or temperature sensors. Herein, we demonstrate a fully transparent and flexible bimodal sensor using indium tin oxide (ITO), which is embedded in a plastic substrate. For the proposed pressure sensor, the embedded ITO is detached from its Mayan-pyramid-structured silicon mold by an environmentally friendly method which utilizes water-soluble sacrificial layers. The Mayan-pyramid-based pressure sensor is capable of six different pressure sensations with excellent sensitivity in the range of 100 Pa-10 kPa, high endurance of 105 cycles, and good pulse detection and tactile sensing data processing capabilities through machine learning (ML) algorithms for different surface textures. A 5 × 5-pixel pressure-temperature-based bimodal sensor array with a zigzag-shaped ITO temperature sensor on top of it is also demonstrated without a noticeable interface effect. This work demonstrates the potential to develop transparent bimodal sensors that can be employed for electronic skin (E-skin) applications.

20.
Cancers (Basel) ; 11(9)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31492002

ABSTRACT

Despite the presence of aggressive treatment strategies, glioblastoma remains intractable, warranting a novel therapeutic modality. An oral antipsychotic agent, penflurido (PFD), used for schizophrenia treatment, has shown an antitumor effect on various types of cancer cells. As glioma sphere-forming cells (GSCs) are known to mediate drug resistance in glioblastoma, and considering that antipsychotics can easily penetrate the blood-brain barrier, we investigated the antitumor effect of PFD on patient-derived GSCs. Using five GSCs, we found that PFD exerts an antiproliferative effect in a time- and dose-dependent manner. At IC50, spheroid size and second-generation spheroid formation were significantly suppressed. Stemness factors, SOX2 and OCT4, were decreased. PFD treatment reduced cancer cell migration and invasion by reducing the Integrin α6 and uPAR levels and suppression of the expression of epithelial-to-mesenchymal transition (EMT) factors, vimentin and Zeb1. GLI1 was found to be involved in PFD-induced EMT inhibition. Furthermore, combinatorial treatment of PFD with temozolomide (TMZ) significantly suppressed tumor growth and prolonged survival in vivo. Immunostaining revealed decreased expression of GLI1, SOX2, and vimentin in the PFD treatment group but not in the TMZ-only treatment group. Therefore, PFD can be effectively repurposed for the treatment of glioblastoma by combining it with TMZ.

SELECTION OF CITATIONS
SEARCH DETAIL
...