Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(12): 822, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38092725

ABSTRACT

Jagged1 (JAG1) is a Notch ligand that correlates with tumor progression. Not limited to its function as a ligand, JAG1 can be cleaved, and its intracellular domain translocates to the nucleus, where it functions as a transcriptional cofactor. Previously, we showed that JAG1 intracellular domain (JICD1) forms a protein complex with DDX17/SMAD3/TGIF2. However, the molecular mechanisms underlying JICD1-mediated tumor aggressiveness remains unclear. Here, we demonstrate that JICD1 enhances the invasive phenotypes of glioblastoma cells by transcriptionally activating epithelial-to-mesenchymal transition (EMT)-related genes, especially TWIST1. The inhibition of TWIST1 reduced JICD1-driven tumor aggressiveness. Although SMAD3 is an important component of transforming growth factor (TGF)-ß signaling, the JICD1/SMAD3 transcriptional complex was shown to govern brain tumor invasion independent of TGF-ß signaling. Moreover, JICD1-TWIST1-MMP2 and MMP9 axes were significantly correlated with clinical outcome of glioblastoma patients. Collectively, we identified the JICD1/SMAD3-TWIST1 axis as a novel inducer of invasive phenotypes in cancer cells.


Subject(s)
Glioblastoma , Humans , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/genetics , Homeodomain Proteins/metabolism , Ligands , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
2.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834227

ABSTRACT

Glioblastoma (GBM) is the most lethal brain cancer, causing inevitable deaths of patients owing to frequent relapses of cancer stem cells (CSCs). The significance of the NOTCH signaling pathway in CSCs has been well recognized; however, there is no NOTCH-selective treatment applicable to patients with GBM. We recently reported that Jagged1 (JAG1), a NOTCH ligand, drives a NOTCH receptor-independent signaling pathway via JAG1 intracellular domain (JICD1) as a crucial signal that renders CSC properties. Therefore, mechanisms regulating the JICD1 signaling pathway should be elucidated to further develop a selective therapeutic regimen. Here, we identified annexin A2 (ANXA2) as an essential modulator to stabilize intrinsically disordered JICD1. The binding of ANXA2 to JICD1 prevents the proteasomal degradation of JICD1 by heat shock protein-70/90 and carboxy-terminus of Hsc70 interacting protein E3 ligase. Furthermore, JICD1-driven propagation and tumor aggressiveness were inhibited by ANXA2 knockdown. Taken together, our findings show that ANXA2 maintains the function of the NOTCH receptor-independent JICD1 signaling pathway by stabilizing JICD1, and the targeted suppression of JICD1-driven CSC properties can be achieved by blocking its interaction with ANXA2.


Subject(s)
Annexin A2 , Glioblastoma , Humans , Annexin A2/genetics , Annexin A2/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Neoplasm Recurrence, Local , Receptors, Notch/metabolism
3.
Biotechnol J ; 17(7): e2100434, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35233982

ABSTRACT

Alternative cancer models that are close to humans are required to create more valuable preclinical results during oncology studies. Here, a new onco-pig model via developing a CRISPR-Cas9-based Conditional Polycistronic gene expression Cassette (CRI-CPC) system to control the tumor inducing simian virus 40 large T antigen (SV40LT) and oncogenic HRASG12V . After conducting somatic cell nuclear transfer (SCNT), transgenic embryos were transplanted into surrogate mothers and five male piglets were born. Umbilical cord analysis confirmed that all piglets were transgenic. Two of them survived and they expressed a detectable green fluorescence. The test was made whether CRI-CPC models were naturally fertile and whether the CRI-CPC system was stably transferred to the offspring. By mating with a normal female pig, four offspring piglets were successfully produced. Among them, only three male piglets were transgenic. Finally, their applicability was tested as cancer models after transduction of Cas9 into fibroblasts from each CRI-CPC pig in vitro, resulting in cell acquisition of cancerous characteristics via the induction of oncogene expression. These results showed that our new CRISPR-Cas9-based onco-pig model was successfully developed.


Subject(s)
CRISPR-Cas Systems , Nuclear Transfer Techniques , Animals , Animals, Genetically Modified , CRISPR-Cas Systems/genetics , Female , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Male , Oncogenes , Swine/genetics
4.
Oncogene ; 39(9): 1969-1982, 2020 02.
Article in English | MEDLINE | ID: mdl-31772327

ABSTRACT

Most of the mechanisms governing apoptotic and non-apoptotic cell death are regulated independently. However, cells may experience various stresses that lead to both apoptotic and non-apoptotic cell death. In particular, cancer cells require a program that simultaneously avoids these forms of cell death, but the mechanism by which they are able to do so is currently unclear. Here, we show that ABC transporter subfamily B member 7 (ABCB7), one of the mitochondrial iron transporters, induces the hypoxia-independent accumulation of hypoxia-inducible factor 1 alpha by controlling intracellular iron homeostasis and inhibits both apoptotic and non-apoptotic cell death. Mechanistically, ABCB7 mitigates non-apoptotic cell death by reducing levels of mitochondrial reactive oxygen species. ABCB7 also suppresses apoptosis by inhibiting the expression of leucine zipper downregulated in cancer 1, an inhibitor of nuclear factor-kappa B signaling. Therefore, our results support that ABCB7 is crucial in controlling both apoptotic and non-apoptotic cell death and indicate that the fine-tuning of intracellular iron homeostasis may be a novel anticancer strategy.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apoptosis , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , ATP-Binding Cassette Transporters/genetics , Biomarkers, Tumor , Cell Death , Cytoplasm/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Iron/metabolism , Mitochondria/genetics , NF-kappa B/genetics , Signal Transduction , Tumor Cells, Cultured
5.
J Ethnopharmacol ; 236: 393-400, 2019 May 23.
Article in English | MEDLINE | ID: mdl-30878548

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Korean Red ginseng extract (RG) is one of the most widely used traditional health functional food in Asia, which invigorates immunity and vital energy. RG have been suggested to inhibit proliferation, invasion, and inflammation in several cancer cell lines. Correspondingly, clinical studies have raised the possibility that RG could augment therapeutic efficacy in cancer patients. However, little is known about the anti-cancer effects of RG in glioblastoma (GBM), the most common and aggressive brain tumor for which effective therapeutic regimens need to be developed. AIM OF THIS STUDY: Here, we assessed the in vivo and in vitro anti-cancer properties of RG in a patient-derived xenograft mouse model and GBM stem cell (GSC) line. MATERIALS AND METHODS: We evaluated the anti-cancer effects of RG in patient-derived GBM xenograft mice with and without combined concurrent chemo- and radiation therapy (CCRT). Furthermore, we verified the in vitro effects of RG on the proliferation, cell death, and stem cell-like self-renewal capacity of cancer cells. Finally, we investigated the signaling pathway affected by RG, via which its anti-cancer effects were mediated. RESULTS: When combined with CCRT, RG impeded GBM progression by reducing cancer cell proliferation and ionized calcium-binding adapter molecule 1 (IBA1)-positive immune cell recruitment. The anti-cancer effects of RG were mediated by Rg3 and Rh2 ginsenosides. Rg3 promoted cell death while Rh2 did not. Furthermore, both Rg3 and Rh2 reduced cell viability and self-renewal capacity of GSCs by inhibiting Wnt/ß-catenin signaling. CONCLUSION: Therefore, our observations imply that RG could be applied to the GBM patients in parallel with CCRT to enhance therapeutic efficacy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Panax/chemistry , Plant Extracts/pharmacology , Adult , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Brain/cytology , Brain/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Glioblastoma/pathology , Humans , Medicine, Korean Traditional , Mice , Neoplastic Stem Cells , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
6.
Cell Death Differ ; 26(10): 2139-2156, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30804471

ABSTRACT

Most glioblastomas frequently recur at sites of radiotherapy, but it is unclear if changes in the tumor microenvironment due to radiotherapy influence glioblastoma recurrence. Here, we demonstrate that radiation-induced senescent glioblastoma cells exhibit a senescence-associated secretory phenotype that functions through NFκB signaling to influence changes in the tumor microenvironment, such as recruitment of Ly6G+ inflammatory cells and vessel formation. In particular, Ly6G+ cells promote conversion of glioblastoma cells to glioblastoma stem cells (GSCs) through the NOS2-NO-ID4 regulatory axis. Specific inhibition of NFκB signaling in irradiated glioma cells using the IκBα super repressor prevents changes in the tumor microenvironment and dedifferentiation of glioblastoma cells. Treatment with Ly6G-neutralizing antibodies also reduces the number of GSCs and prolongs survival in tumor-bearing mice after radiotherapy. Clinically, a positive correlation exists between Ly6G+ cells and the NOS2-NO-ID4 regulatory axis in patients diagnosed with recurrent glioblastoma. Together, our results illustrate important roles for Ly6G+ inflammatory cells recruited by radiation-induced SASP in cancer cell dedifferentiation and tumor recurrence.


Subject(s)
Antigens, Ly/genetics , Brain Neoplasms/genetics , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Transfection
7.
Mol Cells ; 42(2): 135-142, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30622231

ABSTRACT

OCT4, also known as POU5F1 (POU domain class 5 transcription factor 1), is a transcription factor that acts as a master regulator of pluripotency in embryonic stem cells and is one of the reprogramming factors required for generating induced pluripotent stem cells. The human OCT4 encodes three isoforms, OCT4A, OCT4B, and OCT4B1, which are generated by alternative splicing. Currently, the functions and expression patterns of OCT4B remain largely unknown in malignancies, especially in human glioblastomas. Here, we demonstrated the function of OCT4B in human glioblastomas. Among the isoform of OCT4B, OCT4B-190 (OCT4B19kDa) was highly expressed in human glioblastoma stem cells and glioblastoma cells and was mainly detected in the cytoplasm rather than the nucleus. Overexpression of OCT4B19kDa promoted colony formation of glioblastoma cells when grown in soft agar culture conditions. Clinical data analysis revealed that patients with gliomas that expressed OCT4B at high levels had a poorer prognosis than patients with gliomas that expressed OCT4B at low levels. Thus, OCT4B19kDa may play a crucial role in regulating cancer cell survival and adaption in a rigid environment.


Subject(s)
Glioblastoma/metabolism , Glioblastoma/pathology , Octamer Transcription Factor-3/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics , Prognosis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Stress, Physiological , Tumor Stem Cell Assay
8.
Cell Death Differ ; 26(3): 409-425, 2019 03.
Article in English | MEDLINE | ID: mdl-29786075

ABSTRACT

Glioblastoma (GBM), the most severe and common brain tumor in adults, is characterized by multiple somatic mutations and aberrant activation of inflammatory responses. Immune cell infiltration and subsequent inflammation cause tumor growth and resistance to therapy. Somatic loss-of-function mutations in the gene encoding tumor suppressor protein p53 (TP53) are frequently observed in various cancers. However, numerous studies suggest that TP53 regulates malignant phenotypes by gain-of-function (GOF) mutations. Here we demonstrate that a TP53 GOF mutation promotes inflammation in GBM. Ectopic expression of a TP53 GOF mutant induced transcriptomic changes, which resulted in enrichment of gene signatures related to inflammation and chemotaxis. Bioinformatics analyses revealed that a gene signature, upregulated by the TP53 GOF mutation, is associated with progression and shorter overall survival in GBM. We also observed significant correlations between the TP53 GOF mutation signature and inflammation in the clinical database of GBM and other cancers. The TP53 GOF mutant showed upregulated C-C motif chemokine ligand 2 (CCL2) and tumor necrosis factor alpha (TNFA) expression via nuclear factor kappa B (NFκB) signaling, consequently increasing microglia and monocyte-derived immune cell infiltration. Additionally, TP53 GOF mutation and CCL2 and TNFA expression correlated positively with tumor-associated immunity in patients with GBM. Taken together, our findings suggest that the TP53 GOF mutation plays a crucial role in inflammatory responses, thereby deteriorating prognostic outcomes in patients with GBM.


Subject(s)
Brain Neoplasms/genetics , Gain of Function Mutation , Glioblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Genes, p53 , Glioblastoma/pathology , HEK293 Cells , HL-60 Cells , Heterografts , Humans , Inflammation/genetics , Inflammation/pathology , Mice
9.
Biochem Biophys Res Commun ; 496(4): 1013-1018, 2018 02 19.
Article in English | MEDLINE | ID: mdl-28216164

ABSTRACT

Glioma stem-like cells (GSCs) contribute to tumor initiation, progression, and therapeutic resistance, but their cellular origin remains largely unknown. Here, using a stem/progenitor cell-fate tracking reporter system in which eGFP is expressed by promoter of OCT4 that is activated in stem/progenitor cells, we demonstrate that eGFP-negative glioma cells (GCs) became eGFP-positive-GCs in both in vitro cultures and in vivo xenografts. These eGFP-positive-GCs exhibited GSC features and primarily localized to the perivascular region in tumor xenografts, similar to the existence of OCT4-expressing GCs in the perivascular region of human glioblastoma specimens. Angiocrine factors, including nitric oxide (NO), converted eGFP-negative-GCs into eGFP-positive-GCs. Mechanistically, NO signaling conferred GSC features to GCs by increasing OCT4 and NOTCH signaling via ID4. NO signaling blockade and a suicide gene induction prevented tumorigenicity with a decrease in eGFP-positive-GCs in the perivascular region. Taken together, our results reveal the molecular mechanism underlying GSCs generation by cancer cell dedifferentiation.


Subject(s)
Angiogenic Proteins/metabolism , Cell Dedifferentiation , Glioma/metabolism , Glioma/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Mice , Mice, Nude , Neovascularization, Pathologic
10.
Clin Cancer Res ; 24(2): 383-394, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29208670

ABSTRACT

Purpose: Normal stem cells tightly control self-renewal and differentiation during development, but their neoplastic counterparts, cancer stem cells (CSCs), sustain tumorigenicity both through aberrant activation of stemness and evasion of differentiation. Although regulation of CSC stemness has been extensively studied, the molecular mechanisms suppressing differentiation remain unclear.Experimental Design: We performed in silico screening and in vitro validation studies through Western blotting, qRT-PCR for treatment of WNT and SHH signaling inhibitors, and BMP signaling inducer with control and ID1-overexpressing cells. We also performed in vivo drug treatment assays with Balb/c nude mice.Results: Inhibitor of differentiation 1 (ID1) abrogated differentiation signals from bone morphogenetic protein receptor (BMPR) signaling in glioblastoma stem cells (GSCs) to promote self-renewal. ID1 inhibited BMPR2 expression through miRNAs, miR-17 and miR-20a, which are transcriptional targets of MYC. ID1 increases MYC expression by activating WNT and SHH signaling. Combined pharmacologic blockade of WNT and SHH signaling with BMP treatment significantly suppressed GSC self-renewal and extended survival of tumor-bearing mice.Conclusions: Collectively, our results suggested that ID1 simultaneously regulates stemness through WNT and SHH signaling and differentiation through BMPR-mediated differentiation signaling in GSCs, informing a novel therapeutic strategy of combinatorial targeting of stemness and differentiation. Clin Cancer Res; 24(2); 383-94. ©2017 AACR.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Glioma/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein Receptors, Type II/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm , Glioma/genetics , Glioma/pathology , Glioma/therapy , Humans , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Knockout , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Radiation Tolerance , Signal Transduction/drug effects , Transcriptome , Xenograft Model Antitumor Assays
11.
J Med Food ; 20(9): 838-845, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28792781

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most aggressive and lethal human brain tumors, and the median survival of patients with GBM is only 14 months. Glioblastoma stem cells (GSCs) are regarded as a main cause of GBM recurrence, because of their self-renewal and drug resistance properties. Therefore, targeting GSCs is an important therapeutic strategy for GBM. In this study, we show the effects of BRM270, a compound from natural plant extracts, on GSCs in vitro and GBM recurrence in vivo. BRM270 induced apoptotic cell death and inhibited cell growth and "stemness" both in vitro and in vivo. Combining BRM270 treatment with concurrent chemoradiotherapy (CCRT) dramatically increased mice survival and tumor growth inhibition. Taken together, our results suggested that BRM270 synergizes with CCRT as a therapeutic agent to target GSCs.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Glioblastoma/drug therapy , Neoplastic Stem Cells/cytology , Plant Extracts/administration & dosage , Animals , Apoptosis/drug effects , Brain Neoplasms/physiopathology , Brain Neoplasms/radiotherapy , Cell Proliferation/radiation effects , Chemoradiotherapy , Combined Modality Therapy , Glioblastoma/physiopathology , Glioblastoma/radiotherapy , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects
12.
Mol Cells ; 40(7): 515-522, 2017 Jul 31.
Article in English | MEDLINE | ID: mdl-28736425

ABSTRACT

CD133, a pentaspan transmembrane glycoprotein, is generally used as a cancer stem cell marker in various human malignancies, but its biological function in cancer cells, especially in glioma cells, is largely unknown. Here, we demonstrated that forced expression of CD133 increases the expression of IL-1ß and its downstream chemokines, namely, CCL3, CXCL3 and CXCL5, in U87MG glioma cells. Although there were no apparent changes in cell growth and sphere formation in vitro and tumor growth in vivo, in vitro trans-well studies and in vivo tumor xenograft assays showed that neutrophil recruitment was markedly increased by the ectopic expression of CD133. In addition, the clinical relevance between CD133 expression and IL-1ß gene signature was established in patients with malignant gliomas. Thus, these results imply that glioma cells expressing CD133 are capable of modulating tumor microenvironment through the IL-1ß signaling pathway.


Subject(s)
AC133 Antigen/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Interleukin-1beta/metabolism , Neutrophil Infiltration , Signal Transduction , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Chemokines/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Prognosis , Up-Regulation/genetics
13.
Cell Rep ; 16(6): 1629-1641, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477274

ABSTRACT

Inhibitor of differentiation 1 (ID1) is highly expressed in glioblastoma stem cells (GSCs). However, the regulatory mechanism responsible for its role in GSCs is poorly understood. Here, we report that ID1 activates GSC proliferation, self-renewal, and tumorigenicity by suppressing CULLIN3 ubiquitin ligase. ID1 induces cell proliferation through increase of CYCLIN E, a target molecule of CULLIN3. ID1 overexpression or CULLIN3 knockdown confers GSC features and tumorigenicity to murine Ink4a/Arf-deficient astrocytes. Proteomics analysis revealed that CULLIN3 interacts with GLI2 and DVL2 and induces their degradation via ubiquitination. Consistent with ID1 knockdown or CULLIN3 overexpression in human GSCs, pharmacologically combined control of GLI2 and ß-CATENIN effectively diminishes GSC properties. A ID1-high/CULLIN3-low expression signature correlates with a poor patient prognosis, supporting the clinical relevance of this signaling axis. Taken together, a loss of CULLIN3 represents a common signaling node for controlling the activity of intracellular WNT and SHH signaling pathways mediated by ID1.


Subject(s)
Cullin Proteins/metabolism , Glioblastoma/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Hedgehog Proteins/metabolism , Humans , Mice , beta Catenin/metabolism
14.
Tumour Biol ; 37(5): 5857-67, 2016 May.
Article in English | MEDLINE | ID: mdl-26586398

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most aggressive and fatal primary brain tumors in humans. The standard therapy for the treatment of GBM is surgical resection, followed by radiotherapy and/or chemotherapy. However, the frequency of tumor recurrence in GBM patients is very high, and the survival rate remains poor. Delineating the mechanisms of GBM recurrence is essential for therapeutic advances. Here, we demonstrate that irradiation rendered 17-20 % of GBM cells dead, but resulted in 60-80 % of GBM cells growth-arrested with increases in senescence markers, such as senescence-associated beta-galactosidase-positive cells, H3K9me3-positive cells, and p53-p21(CIP1)-positive cells. Moreover, irradiation induced expression of senescence-associated secretory phenotype (SASP) mRNAs and NFκB transcriptional activity in GBM cells. Strikingly, compared to injection of non-irradiated GBM cells into immune-deficient mice, the co-injection of irradiated and non-irradiated GBM cells resulted in faster growth of tumors with the histological features of human GBM. Taken together, our findings suggest that the increases in senescent cells and SASP in GBM cells after irradiation is likely one of main reasons for tumor recurrence in post-radiotherapy GBM patients.


Subject(s)
Cellular Senescence/radiation effects , Glioblastoma/metabolism , Glioblastoma/pathology , Phenotype , Animals , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Disease Models, Animal , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/genetics , Glioblastoma/radiotherapy , Heterografts , Humans , Mice , NF-kappa B/metabolism , Transcriptional Activation
15.
Tumour Biol ; 36(7): 5561-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25680411

ABSTRACT

Interferon regulatory factor 7 (IRF7) is the master transcription factor that plays a pivotal role in the transcriptional activation of type I interferon genes in the inflammatory response. Our previous study revealed that IRF7 is an important regulator of tumor progression via the expression of inflammatory cytokines in glioma. Here, we report that IRF7 promotes glioma invasion and confers resistance to both chemotherapy and radiotherapy by inhibiting expression of argonaute 2 (AGO2), a regulator of microRNA biogenesis. We found that IRF7 and AGO2 expression levels were negatively correlated in patients with glioblastoma multiforme. Ectopic IRF7 expression led to a reduction in AGO2 expression, while depletion of IRF7 resulted in increased AGO2 expression in the LN-229 glioma cell line. In an in vitro invasion assay, IRF7 overexpression enhanced glioma cell invasion. Furthermore, reconstitution of AGO2 expression in IRF7-overexpressing cells led to decreased cell invasion, whereas the reduced invasion due to IRF7 depletion was rescued by AGO2 depletion. In addition, IRF7 induced chemoresistance and radioresistance of glioma cells by diminishing AGO2 expression. Finally, AGO2 depletion alone was sufficient to accelerate glioma cell invasion in vitro and in vivo, indicating that AGO2 regulates cancer cell invasion. Taken together, our results indicate that IRF7 promotes glioma cell invasion and both chemoresistance and radioresistance through AGO2 inhibition.


Subject(s)
Argonaute Proteins/biosynthesis , Brain Neoplasms/genetics , Glioma/genetics , Interferon Regulatory Factor-7/biosynthesis , Argonaute Proteins/antagonists & inhibitors , Argonaute Proteins/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/pathology , Glioma/radiotherapy , Humans , Interferon Regulatory Factor-7/genetics , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Radiation Tolerance/genetics
16.
Arch Pharm Res ; 38(3): 389-401, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25502807

ABSTRACT

Chemo-resistance and radio-resistance are a major cause of recurrence and progression of many cancers, regardless of improvements in therapies. Since cancer stem cells (CSCs) were identified as a rare population with the abilities of self-renewal; tumor initiation; aberrant differentiation, which contributes to tumor heterogeneity; and resistance to anticancer therapeutics, they have been considered a major cause of tumor recurrence post-therapy and a primary therapeutic target in relapse prevention. A number of studies have demonstrated the mechanisms underlying chemo-resistance and radio-resistance of CSCs. In this review, we describe intrinsic and extrinsic factors underlying CSC chemo-resistance and radio-resistance. The intrinsic factors regulate CSC signaling pathways involved in stem cell signaling, anti-apoptotic pathways, ABC transporter expression, and DNA damage repair systems. The extrinsic factors include the resistance mechanisms resulting from the interactions between CSCs and the microenvironment composed of vessels, fibroblasts, immune cells, extracellular matrix, and diverse soluble factors. Furthermore, we introduce diverse therapeutic agents used in experimental or clinical trials to target CSCs. Understanding how CSCs acquire resistance to anticancer therapeutics will give us opportunity to develop improved therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Disease Progression , Drug Resistance, Neoplasm , Humans , Neoplasm Recurrence, Local , Neoplasms/pathology , Radiation Tolerance
17.
Cancer Lett ; 353(2): 194-200, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25079688

ABSTRACT

The invasiveness of glioblastoma is a major cause of poor prognosis and relapse. However, the molecular mechanism controlling glioma cell invasion is poorly understood. Here, we report that receptor activator of nuclear factor kappa-B (NFκB) ligand (RANKL) promotes glioma cell invasion in vivo, but not in vitro. Unlike the invasiveness under in vitro culture conditions, in vivo xenograft studies revealed that LN229 cells expressing high endogenous RANKL generated more invasive tumors than U87MG cells expressing relatively low endogenous RANKL. Consistently, RANKL-overexpressing U87MG resulted in invasive tumors, whereas RANKL-depleted LN229 generated rarely invasive tumors. We found that the number of activated astrocytes was markedly increased in the periphery of RANKL-high invasive tumors. RANKL activated astrocytes through NFκB signaling and these astrocytes in turn secreted various factors which regulate glioma cell invasion. Among them, transforming growth factor ß (TGF-ß) signaling was markedly increased in glioblastoma specimens and xenograft tumors expressing high levels of RANKL. These results indicate that RANKL contributes to glioma invasion by modulating the peripheral microenvironment of the tumor, and that targeting RANKL signaling has important implications for the prevention of highly invasive glioblastoma.


Subject(s)
Astrocytes/physiology , Brain Neoplasms/metabolism , Cytokines/physiology , Glioblastoma/metabolism , RANK Ligand/physiology , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Signal Transduction , Tumor Microenvironment
18.
Mol Biol Rep ; 41(9): 5903-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24965146

ABSTRACT

The Rab protein family is composed of small GTP-binding proteins involved in intracellular vesicle trafficking. In particular, Rab3a which is one of four Rab3 proteins (a, b, c, and d isoforms) is associated with synaptic vesicle trafficking in normal brain. However, despite the elevated level of Rab3a in tumors, its role remains unclear. Here we report a tumorigenic role of Rab3a in brain tumors. Elevated level of Rab3a expression in human was confirmed in both glioma cell lines and glioblastoma multiforme patient specimens. Ectopic Rab3a expression in glioma cell lines and primary astrocytes promoted cell proliferation by increasing cyclin D1 expression, induced resistance to anti-cancer drug and irradiation, and accelerated foci formation in soft agar and tumor formation in nude mice. The overexpression of Rab3a augmented the tumorsphere-forming ability of glioma cells and p53(-/-) astrocytes and increased expression levels of various stem cell markers. Taken together, our results indicate that Rab3a is a novel oncogene involved in glioma initiation and progression.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , rab3A GTP-Binding Protein/metabolism , Animals , Astrocytes/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Progression , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Humans , Mice , Mice, Knockout , Mice, Nude , rab3A GTP-Binding Protein/genetics
19.
Brain ; 135(Pt 4): 1055-69, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22434214

ABSTRACT

Inflammatory microenvironment signalling plays a crucial role in tumour progression (i.e. cancer cell proliferation, survival, angiogenesis and metastasis) in many types of human malignancies. However, the role of inflammation in brain tumour pathology remains poorly understood. Here, we report that interferon regulatory factor 7 is a crucial regulator of brain tumour progression and heterogeneity. Ectopic expression of interferon regulatory factor 7 in glioma cells promotes tumorigenicity, angiogenesis, microglia recruitment and cancer stemness in vivo and in vitro through induction of interleukin 6, C-X-C motif chemokine 1 and C-C motif chemokine 2. In particular, interferon regulatory factor 7-driven interleukin 6 plays a pivotal role in maintaining glioma stem cell properties via Janus kinase/signal transducer and activator of transcription-mediated activation of Jagged-Notch signalling in glioma cells and glioma stem cells derived from glioma patients. Accordingly, the short hairpin RNA-mediated depletion of interferon regulatory factor 7 in glioma stem cells markedly suppressed interleukin 6-Janus kinase/signal transducer and activator of transcription-mediated Jagged-Notch-signalling pathway, leading to decreases in glioma stem cell marker expression, tumoursphere-forming ability, and tumorigenicity. Furthermore, in a mouse model of wound healing, depletion of interferon regulatory factor 7 suppressed tumour progression and decreased cellular heterogeneity. Finally, interferon regulatory factor 7 was overexpressed in patients with high-grade gliomas, suggesting its potential as an independent prognostic marker for glioma progression. Taken together, our findings indicate that interferon regulatory factor 7-mediated inflammatory signalling acts as a major driver of brain tumour progression and cellular heterogeneity via induction of glioma stem cell genesis and angiogenesis.


Subject(s)
Glioma/pathology , Interferon Regulatory Factor-7/metabolism , Interleukin-6/metabolism , Neoplastic Stem Cells/physiology , Receptor, Notch1/metabolism , Signal Transduction/physiology , AC133 Antigen , Antigens, CD/metabolism , Astrocytes/metabolism , Brain/cytology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Chromatin Immunoprecipitation , Computational Biology , Endothelial Cells , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glycoproteins/metabolism , Humans , Interferon Regulatory Factor-7/genetics , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Peptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transduction, Genetic/methods , Tumor Stem Cell Assay
20.
Cancer Res ; 71(8): 3066-75, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21363911

ABSTRACT

One of the most detrimental hallmarks of glioblastoma multiforme (GBM) is cellular invasiveness, which is considered a potential cause of tumor recurrence. Infiltrated GBM cells are difficult to completely eradicate surgically and with local therapeutic modalities. Although much effort has focused on understanding the various mechanisms controlling GBM invasiveness, its nature remains poorly understood. In this study, we established highly serial intracranial transplantation. U87R4 cells were highly invasive and displayed stem cell-like properties, as compared to noninvasive but proliferative U87L4 cells. Microarray analysis during serial transplantation revealed that apoptosis-inducing genes (caspase3 and PDCD4) were downregulated whereas several cancer stem cell-relevant genes [Frizzled 4 (FZD4) and CD44] were upregulated in more invasive cells. U87R4 cells were resistant to anticancer drug-induced cell death, partly due to downregulation of caspase3 and PDCD4, and they retained activated Wnt/ß-catenin signaling due to upregulation of Frizzled 4, which was sufficient to control neurosphere formation. We also found that FZD4 promoted expression of the epithelial to mesenchymal transition regulator SNAI1, along with acquisition of a mesenchymal phenotype. Taken together, our results argue that Frizzled 4 is a member of the Wnt signaling family that governs both stemness and invasiveness of glioma stem cells, and that it may be a major cause of GBM recurrence and poor prognosis.


Subject(s)
Brain Neoplasms/pathology , Frizzled Receptors/biosynthesis , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/biosynthesis , Animals , Apoptosis Regulatory Proteins/biosynthesis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Caspase 3/biosynthesis , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , RNA-Binding Proteins/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...