Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Sci ; 112(9): 3756-3768, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34145699

ABSTRACT

High expression of gangliosides GD3 and GD2 is observed in human gliomas. The functions of GD3 and GD2 in malignant properties have been reported in glioma cells in vitro, but those functions have not yet been investigated in vivo. In this study, we showed that deficiency of GD3 synthase (GD3S, St8sia1) attenuated glioma progression and clinical and pathological features in a platelet-derived growth factor B-driven murine glioma model. Lack of GD3S resulted in the prolonged lifespan of glioma-bearing mice and low-grade pathology in generated gliomas. Correspondingly, they showed reduced phosphorylation levels of Akt, Erks, and Src family kinases in glioma tissues. A DNA microarray study revealed marked alteration in the expression of various genes, particularly in MMP family genes, in GD3S-deficient gliomas. Re-expression of GD3S restored expression of MMP9 in primary-cultured glioma cells. We also identified a transcription factor, Ap2α, expressed in parallel with GD3S expression, and showed that Ap2α was critical for the induction of MMP9 by transfection of its cDNA and luciferase reporter genes, and a ChIP assay. These findings suggest that GD3S enhances the progression of gliomas by enhancement of the Ap2α-MMP9 axis. This is the first report to describe the tumor-enhancing functions of GD3S in vivo.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Disease Models, Animal , Glioma/genetics , Glioma/pathology , Sialyltransferases/genetics , Animals , Astrocytes/metabolism , Cells, Cultured , Disease Progression , Gangliosides/metabolism , Gene Expression Regulation, Neoplastic , Longevity/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transfection
2.
J Pathol ; 243(4): 468-480, 2017 12.
Article in English | MEDLINE | ID: mdl-28888050

ABSTRACT

In the progression of glioma, tumour cells often exploit the perivascular microenvironment to promote their survival and resistance to conventional therapies. Some of these cells are considered to be brain tumour stem cells (BTSCs); however, the molecular nature of perivascular tumour cells has not been specifically clarified because of the complexity of glioma. Here, we identified CD109, a glycosylphosphatidylinositol-anchored protein and regulator of multiple signalling pathways, as a critical regulator of the progression of lower-grade glioma (World Health Organization grade II/III) by clinicopathological and whole-genome sequencing analysis of tissues from human glioma. The importance of CD109-positive perivascular tumour cells was confirmed not only in human lower-grade glioma tissues but also in a mouse model that recapitulated human glioma. Intriguingly, BTSCs isolated from mouse glioma expressed high levels of CD109. CD109-positive BTSCs exerted a proliferative effect on differentiated glioma cells treated with temozolomide. These data reveal the significance of tumour cells that populate perivascular regions during glioma progression, and indicate that CD109 is a potential therapeutic target for the disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Antigens, CD/genetics , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Disease Progression , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Knockout , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Signal Transduction , Temozolomide , Time Factors , Tumor Cells, Cultured , Tumor Microenvironment
3.
J Biol Chem ; 290(26): 16043-58, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940087

ABSTRACT

There have been a few studies on the ganglioside expression in human glioma tissues. However, the role of these gangliosides such as GD3 and GD2 has not been well understood. In this study we employed a genetically engineered mouse model of glioma to clarify the functions of GD3 in gliomas. Forced expression of platelet-derived growth factor B in cultured astrocytes derived from p53-deficient mice resulted in the expression of GD3 and GD2. GD3-positive astrocytes exhibited increased cell growth and invasion activities along with elevated phosphorylation of Akt and Yes kinase. By enzyme-mediated activation of radical sources reaction and mass spectrometry, we identified PDGF receptor α (PDGFRα) as a GD3-associated molecule. GD3-positive astrocytes showed a significant amount of PDGFRα in glycolipid-enriched microdomains/rafts compared with GD3-negative cells. Src kinase family Yes was co-precipitated with PDGFRα, and its pivotal role in the increased cell invasion of GD3-positive astrocytes was demonstrated by silencing with anti-Yes siRNA. Direct association between PDGFRα and GD3 was also shown, suggesting that GD3 forms ternary complex with PDGFRα and Yes. The fact that GD3, PDGFRα, and activated Yes were colocalized in lamellipodia and the edge of tumors in cultured cells and glioma tissues, respectively, suggests that GD3 induced by platelet-derived growth factor B enhances PDGF signals in glycolipid-enriched microdomain/rafts, leading to the promotion of malignant phenotypes such as cell proliferation and invasion in gliomas.


Subject(s)
Brain Neoplasms/metabolism , Gangliosides/metabolism , Glioma/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Glioma/enzymology , Glioma/genetics , Humans , Mice , Neoplasm Invasiveness , Protein Binding , Proto-Oncogene Proteins c-yes/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics
4.
Cancer Sci ; 107(10): 1477-1483, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27467121

ABSTRACT

Sonidegib is a selective inhibitor of Smoothened receptor, which is a key regulator of the Hedgehog signaling pathway. The purpose of this study was to determine the maximum tolerated dose based on dose-limiting toxicity (DLT) and the recommended dose (RD) of sonidegib in Asian patients with advanced solid tumors. This was an open-label, single-arm, multicenter, two-group, parallel, dose-escalation, phase I study undertaken in Asian patients; group 1 included patients from Japan and group 2 included patients from Hong Kong and Taiwan. Dose escalation was guided by a Bayesian logistic regression model dependent on DLTs in cycle 1 and other safety findings. A total of 45 adult Asian patients with confirmed advanced solid tumors were enrolled. Group 1 included 21 patients (12 treated with 400 mg q.d. [once daily] and 9 treated with 600 mg q.d.) and group 2 included 24 patients (12 treated with 400 mg q.d., 8 treated with 600 mg q.d., and 4 treated with 800 mg q.d.). Elevation in creatine kinase was the DLT in both groups. The most common adverse events suspected to be related to sonidegib in both patient groups were increase in creatine kinase levels, myalgia, fatigue, and abnormal hepatic function. The RD of 400 mg q.d. was defined in both groups. Difference in tolerability was noted between the East Asian patients and Western population. The RD in East Asian patients (400 mg q.d.) was lower than in patients from Europe and the USA (800 mg q.d. and 250 mg twice daily). (Registered with Clinicaltrials.gov: NCT01208831.).


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Pyridines/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Asian People , Biomarkers , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/mortality , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Treatment Outcome , Young Adult
5.
Neuropathology ; 34(5): 484-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24738632

ABSTRACT

Papillary glioneuronal tumor (PGNT) is a rare type of primary brain tumor. Although PGNT has traditionally been defined as a clinically indolent neoplasm, several cases with high proliferative activity and tumor recurrence have recently been reported. We report a case of PGNT in a 12-year-old boy who presented with epilepsy and harbored a 64 mm cystic tumor with a high proliferative component in the right temporal lobe. (11) C-methionine positron emission tomography (PET) showed high uptake in the solid mass. Gross total resection of the tumor mass was achieved and the patient became seizure-free without any neurological deficits. Histologically, the tumor contained two distinct areas of a vasocentric papilliform structure and a desmoplastic component. Minigemistocytic cells and small necrotic regions were observed adjacent to the pseudopapillae. Immunohistochemical analyses revealed both glial and neuronal differentiation. The Ki-67 proliferation index was high (14%) in the area corresponding to the high uptake region in the (11) C-methionine PET. No tumor recurrence was observed 20 months after surgery. High proliferative PGNTs are rare and to our knowledge this is only the third pediatric case of PGNT with atypical features reported in the literature. Hence, we here review the reported cases of PGNT and discuss the clinical, radiological and histological features of this malignancy.


Subject(s)
Brain Neoplasms/pathology , Neoplasms, Complex and Mixed/pathology , Astrocytoma/diagnostic imaging , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Carbon Radioisotopes , Carcinoma, Papillary/diagnostic imaging , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Proliferation , Child , Humans , Magnetic Resonance Imaging , Male , Neoplasms, Complex and Mixed/diagnostic imaging , Neoplasms, Complex and Mixed/metabolism , Positron-Emission Tomography
6.
Cancer Sci ; 103(10): 1871-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22747609

ABSTRACT

Recent gene expression and copy number profilings of glioblastoma multiforme (GBM) by The Cancer Genome Atlas (TCGA) Research Network suggest the existence of distinct subtypes of this tumor. However, these approaches might not be easily applicable in routine clinical practice. In the current study, we aimed to establish a proteomics-based subclassification of GBM by integrating their genomic and epigenomic profiles. We subclassified 79 newly diagnosed GBM based on expression patterns determined by comprehensive immunohistochemical observation in combination with their DNA copy number and DNA methylation patterns. The clinical relevance of our classification was independently validated in TCGA datasets. Consensus clustering identified the four distinct GBM subtypes: Oligodendrocyte Precursor (OPC) type, Differentiated Oligodendrocyte (DOC) type, Astrocytic Mesenchymal (AsMes) type and Mixed type. The OPC type was characterized by highly positive scores of Olig2, PDGFRA, p16, p53 and synaptophysin. In contrast, the AsMes type was strongly associated with strong expressions of nestin, CD44 and podoplanin, with a high glial fibrillary acidic protein score. The median overall survival of OPC-type patients was significantly longer than that of the AsMes-type patients (19.9 vs 12.8 months). This finding was in agreement with the Oncomine analysis of TCGA datasets, which revealed that PDGFRA and Olig2 were favorable prognostic factors and podoplanin and CD44 were associated with a poor clinical outcome. This is the first study to establish a subclassification of GBM on the basis of immunohistochemical analysis. Our study will shed light on personalized therapies that might be feasible in daily neuropathological practice.


Subject(s)
Brain Neoplasms/classification , Glioblastoma/classification , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cluster Analysis , Female , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Multiplex Polymerase Chain Reaction , Proteomics , Transcriptome , Young Adult
7.
Nagoya J Med Sci ; 74(3-4): 353-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23092108

ABSTRACT

A 67-year-old woman presented with an acute onset of left-sided weakness. Magnetic resonance (MR) imaging revealed multiple cerebral infarctions and gadolinium-enhanced lesions in both cerebral hemispheres. Her symptoms once improved after starting steroid treatment; however, soon developed consciousness disturbance and hemiparesis on the left side. She was referred to our hospital where she underwent stereotactic needle biopsy, that revealed an intravascular large B-cell lymphoma in the cerebrum. She received high-dose methotrexate chemotherapy followed by whole-brain radiation therapy, and the MR findings improved. However, her medical condition gradually worsened, and she died 6 months after disease onset. Intravascular lymphoma (IVL) limited to the central nervous system (CNS) is very rare, and the optimal treatment for this medical condition has not been established yet. IVLs showing only neurologic manifestations might be overlooked or misdiagnosed as cerebral infarctions. Here, we present a case of CNS IVL, with its radiographic and pathologic features and treatment with high-dose methotrexate chemotherapy.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Cerebral Infarction/diagnosis , Lymphoma/diagnosis , Aged , Female , Humans
8.
Nagoya J Med Sci ; 83(3): 535-549, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34552288

ABSTRACT

Ganglioside GD3/GD2 are over-expressed in various neuroectoderm-derived tumors. Previous studies indicated that GD3 is involved in the enhancement of cancer properties such as rapid growth and increased invasiveness. However, little is known about the functions of GD3/GD2 in glioma cells and glioma microenvironments. To clarify the functions of GD3/GD2 in gliomas, we used a mouse glioma model based on the RCAS/Gtv-a system. At first, we compared the gliomas size between wild-type (WT) and GD3 synthase (GD3S) knockout (KO) mice, showing a less malignant histology and slower tumor growth in GD3S-KO mice than in WT mice. Immunohistochemistry of glioma sections from WT and GD3S-KO mice revealed that reactive microglia/macrophages showed different localization patterns between the two genetic types of mice. CD68+ cells were more frequently stained inside glioma tissues of GD3S-KO mice, while they were stained mainly around glioma tissues in WT mice. The number of CD68+ cells markedly increased in tumor tissues of GD3S-KO mice at 2 weeks after injection of transfectant DF-1 cells. Furthermore, CD68+ cells in GD3S(-/-) glioma tissues expressed higher levels of inducible nitric oxide synthase. We observed higher expression levels of pro-inflammatory cytokine genes in primary-cultured glioma cells of WT mice than in GD3S-KO mice. DNA microarray data also revealed differential expression levels of various cytokines and chemokines in glioma tissues between WT and GD3S-KO mice. These results suggest that expression of GD3S allows glioma cells to promote polarization of microglia/macrophages towards M2-like phenotypes by modulating the expression levels of chemokines and cytokines.


Subject(s)
Glioma , Animals , Cytokines , Glioma/genetics , Mice , Mice, Knockout , Severity of Illness Index , Tumor Microenvironment
9.
J Neurooncol ; 99(1): 147-53, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20063176

ABSTRACT

The simultaneous occurrence of multiple primary intracranial tumors has been reported previously. However, most of these tumors arise after cranial radiotherapy or in association with familial tumor syndromes. Double tumors of different histologies that are unrelated to radiotherapy or genetic disorders are very rare. We present a case of two primary intracranial tumors occurring simultaneously at adjacent sites. Preoperative gadolinium-enhanced magnetic resonance imaging of these tumors revealed a single continuous lesion. Postoperative histological examination revealed the presence of two distinct tumors, meningioma and glioblastoma multiforme. To elucidate the mechanism of synchronous tumor formation, we performed immunohistochemical analysis of the proteins involved in the receptor tyrosine kinase, Wnt, and Notch signaling pathways. These analyses showed that platelet-derived growth factor (PDGF) receptors-alpha and beta were overexpressed in both tumors, thereby indicating the oncogenic effects of activated signaling of these receptors. The PDGF-mediated paracrine system may induce one tumor from another.


Subject(s)
Brain Neoplasms/complications , Glioblastoma/complications , Meningeal Neoplasms/complications , Meningioma/complications , Aged , Brain Neoplasms/diagnosis , Coronary Angiography/methods , Female , Fluorodeoxyglucose F18 , Glioblastoma/diagnosis , Humans , Magnetic Resonance Imaging/methods , Meningeal Neoplasms/pathology , Meningioma/diagnosis , Positron-Emission Tomography/methods
10.
J Neurooncol ; 98(3): 341-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20012911

ABSTRACT

Several biomarkers have been identified as prognostic factors in primary central nervous system lymphoma (PCNSL). However, the correlation between the histogenetic origin of PCNSL and the response to therapy is still unclear. To elucidate the utility of immunophenotypic markers in predicting clinical outcomes, we investigated 27 immunocompetent patients with PCNSL treated with high-dose methotrexate therapy. Of the 27 patients, 25 received whole-brain radiotherapy after high-dose methotrexate. Immunostaining for CD5, CD10, BCL-6, and MUM-1 was used to determine the immunophenotypic profile of diffuse large B-cell lymphoma of PCNSL. We then evaluated whether immunophenotypic markers were associated with the response to therapy or patients' survival. The response to induction high-dose methotrexate therapy was determined by magnetic resonance imaging after three courses of i.v. high-dose methotrexate. We categorized B-cell lymphomas into three known subtypes: germinal center B-cell (GCB), activated-GCB, and post-GCB subtypes according to immunohistochemical profile. All the BCL-6-positive samples were co-positive for MUM-1 and therefore classified into activated-GCB subtype. BCL-6 expression in this study was associated with poor progression-free survival (P = 0.038). No immunophenotypic markers or subtypes had a significant effect on the response to high-dose methotrexate therapy. However, the response itself was a significant predictor for both progression-free survival (P < 0.001) and overall survival (P = 0.003). Further investigation is needed to assess BCL-6 as a potential prognostic factor in PCNSL.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/drug therapy , Immunohistochemistry/methods , Immunosuppressive Agents/therapeutic use , Lymphoma/diagnosis , Lymphoma/drug therapy , Methotrexate/therapeutic use , Adult , Aged , Antigens, CD/metabolism , Central Nervous System Neoplasms/classification , DNA-Binding Proteins/metabolism , Disease-Free Survival , Female , Follow-Up Studies , Humans , Interferon Regulatory Factors/metabolism , Lymphoma/classification , Male , Middle Aged , Proto-Oncogene Proteins c-bcl-6 , Survival Analysis , Time Factors
11.
Pediatr Blood Cancer ; 55(3): 577-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20658636

ABSTRACT

We present a case of a 12-year-old female with a germline TP53 mutation who presented with anaplastic astrocytoma and subsequent acute lymphoblastic leukemia (ALL) 13 months after starting treatment with temozolomide (TMZ). The patient had no family history of malignancy except her grand father and his siblings. Although alkylating agents such as TMZ are known to induce secondary hematologic malignancy, only several cases of treatment-related acute leukemia have been reported after TMZ-alone chemotherapy for malignant gliomas. We demonstrate a rare case of TMZ-related ALL in a child with glioma possibly associated with a germline TP53 mutation.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Frontal Lobe , Genes, p53/genetics , Germ-Line Mutation , Neoplasms, Second Primary/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Antineoplastic Agents, Alkylating/therapeutic use , Astrocytoma/genetics , Brain Neoplasms/genetics , Child , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Female , Humans , Temozolomide
13.
No Shinkei Geka ; 37(10): 973-81, 2009 Oct.
Article in Japanese | MEDLINE | ID: mdl-19882957

ABSTRACT

OBJECTIVES: Patients with malignant gliomas have an even worse prognosis than other cancer patients, and they sometimes undergo surgery and chemo-radiotherapy without having been informed of the nature of the disease and its prognosis in Japan. Often, patients with glioblastoma are only told that they have a brain tumor, although other family members are told of the real diagnosis and prognosis. Since patients with glioblastoma, often experience a rapid deterioration in their condition, they usually do not have enough time to seek a second opinion regarding their disease. We surveyed neurosurgeons in Japan with regard to their policies and attitudes concerning the disclosure of a diagnosis of glioma and their thoughts on the end-of-life care of glioma patients. METHODS: A survey was performed in November, 2007. A questionnaire was sent by e-mail to 259 participants who planned to attend the 25th Brain Tumor Conference in Japan. RESULTS: One hundred and thirty-two participants (51%) returned the questionnaire. Almost all the respondents were neurosurgeons specializing in malignant brain tumors. The percentages of respondents who informed their patients of a diagnosis of grade 2 astrocytoma, grade 3 or grade 4 glioblastoma were 73%, 57% and 37%, respectively. More than 80% of all glioblastoma patients were only told that they had a malignant brain tumor. Sixty-eight percent of the doctors told the family members, but not the patients, of the real diagnosis. The neurosurgeons' policies and attitudes toward end-of-life care for patients with gliomas were also analyzed. CONCLUSIONS: Most neurosurgeons have difficulty disclosing a diagnosis of glioma and providing end-of-life care. This survey will help to develop guidelines regarding disclosure and the decision to pursue end-of-life care for patients with gliomas. (Received : March 19, 2009, Accepted : June 8, 2009)


Subject(s)
Attitude of Health Personnel , Brain Neoplasms/therapy , Glioma/therapy , Neurosurgery , Truth Disclosure , Data Collection , Humans , Japan , Terminal Care
14.
Neuro Oncol ; 21(8): 993-1004, 2019 08 05.
Article in English | MEDLINE | ID: mdl-30976792

ABSTRACT

BACKGROUND: Embryonal tumors in the central nervous system (CNS) are primary, aggressive, and poorly differentiated pediatric brain tumors. We identified forkhead box R2 (Foxr2) as an oncogene for medulloblastoma through a transposon-based insertional mutagenesis screen. Foxr2 translocation has been identified in a subset of human embryonal tumors of the CNS, designated as CNS neuroblastoma with Foxr2 activation (CNS NB-Foxr2); however, the in vivo functions of Foxr2 remain elusive. METHODS: We analyzed the effect of Foxr2 overexpression in the mouse brain by generating a transgenic strain that expresses Foxr2 in the entire brain under a transformation related protein 53 (Trp53)-deficient background. We performed histological analysis of tumors and characterized tumor-derived sphere-forming cells. We investigated gene expression profiles of tumor-derived cells. RESULTS: Foxr2 and Trp53 loss promoted tumor formation in the olfactory bulb (OB) and brainstem (BS). The tumors showed the common morphological features of small round blue cell tumors, exhibiting divergent, mainly neuronal and glial, patterns of differentiation, which corresponds to the definition of CNS-embryonal tumors. Importantly, all mice developed CNS-embryonal tumors. In the OB, early proliferative lesions consisting of oligodendrocyte transcription factor 2 (Olig2+) cells were observed, indicating that Foxr2 expression expanded Olig2+ cells in the OB. Tumor-derived cells formed spheres in vitro and induced tumors that recapitulated the parental tumor upon transplantation, indicating the presence of tumor-initiating cells. Gene expression profiling revealed that OB and BS tumor cells were enriched for the expression of the genes specific to CNS NB-Foxr2. CONCLUSION: Our data demonstrate that Foxr2 plays a causative role in the formation of CNS-embryonal tumors.


Subject(s)
Central Nervous System Neoplasms , Cerebellar Neoplasms , Forkhead Transcription Factors/genetics , Medulloblastoma , Neoplasms, Germ Cell and Embryonal , Animals , Mice
15.
Int J Oncol ; 52(4): 1255-1266, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29436609

ABSTRACT

Sialic acid-containing glycosphingolipids, gangliosides, are considered as cancer associated antigens in neuro-ectoderm-derived tumors such as melanomas and neuroblastomas. In particular, gangliosides GD3 and GD2 are expressed in human gliomas. It has been reported that their expression levels increase along with increased malignant properties. However, the implication of GD3/GD2 in human glioma cells has never been clarified, at least to the best of our knowledge. In this study, we introduced the cDNA of GD3 synthase (GD3S)(ST8SIA1) into a glioma cell line, U-251MG, that expresses neither GD3 nor GD2, thereby establishing transfectant cells U-251MG-GD3S(+) expressing high levels of GD3 and GD2 on the cell surface. In these U-251MG­GD3S(+) cell lines, signaling molecules such as Erk1/2, Akt, p130Cas, paxillin and focal adhesion kinase were activated, leading to the enhancement of invasion activity and motility. It was then demonstrated that the U-251MG-GD3S(+) cells could proliferate under culture conditions with low or no serum concentrations without undergoing cell cycle arrest by escaping the accumulation of p16 and p21. All these results suggested that GD3 and GD2 highly expressed in gliomas confer increased invasion and mobility, cell growth abilities under low serum conditions, and increased ratios of the S-G2/M phase in the cell cycle.


Subject(s)
Brain Neoplasms/pathology , Gangliosides/metabolism , Glioma/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Glioma/metabolism , Humans , Neoplasm Invasiveness/pathology , Tumor Cells, Cultured
16.
Cancer Res ; 65(16): 7429-35, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103096

ABSTRACT

Perifosine is an oral Akt inhibitor which exerts a marked cytotoxic effect on human tumor cell lines, and is currently being tested in several phase II trials for treatment of major human cancers. However, the efficacy of perifosine in human gliomas has not been established. As Akt is activated in approximately 70% of human glioblastomas, we investigated the impact of perifosine on glia in culture and on a mouse glioma model in vivo. Here we show that perifosine strongly reduces phosphorylation levels of Akt and extracellular signal-regulated kinase (Erk) 1/2, induces cell cycle arrest in G1 and G2, and causes dose-dependent growth inhibition of mouse glial progenitors in which Akt and/or Ras-Erk 1/2 pathways are activated. Furthermore, because temozolomide is a common oral alkylating agent used in the treatment of gliomas, we investigated the effect of perifosine in combination with temozolomide. We observed an enhanced effect when both were used in culture. With these results, we combined perifosine and temozolomide as treatment of platelet-derived growth factor B-driven gliomas in mice. Animal studies showed that perifosine and temozolomide combination therapy was more effective than temozolomide treatment alone (P < 0.01). These results indicate that perifosine is an effective drug in gliomas in which Akt and Ras-Erk 1/2 pathways are frequently activated, and may be a new candidate for glioma treatment in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Phosphorylcholine/analogs & derivatives , Animals , Brain Neoplasms/pathology , CDC2-CDC28 Kinases/antagonists & inhibitors , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase 2 , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , G1 Phase/drug effects , G2 Phase/drug effects , Glioma/pathology , Humans , Mice , Mice, Transgenic , Phosphorylcholine/pharmacology , Platelet-Derived Growth Factor/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Temozolomide , Xenograft Model Antitumor Assays
17.
Curr Opin Biotechnol ; 16(6): 681-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16263256

ABSTRACT

As continuous cell proliferation caused by genetic alterations leads to cancer, monitoring abnormal cell proliferation in sporadic tumor models is important in the context of tumor generation, development and response to therapy. Bioluminescence imaging technology, which visualizes the conversion of chemical energy into visible light by luciferase enzymes, is an established method to measure cell numbers in grafted tumors in vivo, but has not been used to monitor cell proliferation per se. To measure cell proliferation noninvasively, transgenic mice have been developed that express the luciferase gene under the control of the E2F1 promoter. When these reporter mice are crossed with genetically defined mouse models of human cancer, the proliferative activity of the tumor cells can be monitored with proportional light production. These technologies support more detailed preclinical trials and could enable other biological pathways to be monitored in living cells.


Subject(s)
Cell Proliferation , Diagnostic Imaging , E2F Transcription Factors/genetics , Luciferases/biosynthesis , Luminescent Measurements , Neoplasms, Experimental/metabolism , Animals , Diagnostic Imaging/methods , Diagnostic Imaging/trends , Gene Transfer Techniques , Humans , Luciferases/genetics , Luminescent Measurements/methods , Luminescent Measurements/trends , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics
18.
Brain Tumor Pathol ; 32(4): 281-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26334755

ABSTRACT

Primary peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), is a rare disease that infrequently involves the central nervous system (CNS), and it is even rarer in pediatric patients. Here, we report of a 13-year-old male with primary CNS PTCL-NOS who exhibited a malignant clinical course with recurrence after radiochemotherapy followed by bone marrow transplantation; he died 43 months after diagnosis. Pathology revealed the proliferation of cytotoxic T-cells and clonal T-cell receptor gene rearrangements. Although the optimal therapy for PTCL remains controversial, intensive radiochemotherapy may be required for some patients.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/therapy , Adolescent , Bone Marrow Transplantation , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Fatal Outcome , Gene Rearrangement , Genes, T-Cell Receptor/genetics , Humans , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/pathology , Male , Neoplasm Recurrence, Local , Radiochemistry
19.
Brain Tumor Pathol ; 32(2): 137-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24807103

ABSTRACT

Meningiomas occasionally display aggressive behavior, but the mechanisms of malignant transformation remain unclear. We encountered the case of a 65-year-old man with a 10-year history of recurrent meningioma. The patient had undergone multiple tumor resections, radiotherapy treatments, and reconstructive surgeries due to wound infection. After the third resection of the tumor and reconstruction with an omental flap, the tumor demonstrated rapid growth and lung metastasis. The final pathological diagnosis was anaplastic meningioma. Because the drastic change of the tumor was observed after omental flap transposition, we investigated the effect of the omentum on tumor cells and performed histopathological analyses of meningiomas using a mouse model. We found that meningioma cells have a high affinity to the omentum and show a growth advantage when co-cultured with adipocytes. Immunohistochemical staining revealed that meningioma cells adjacent to the omentum strongly expressed fatty acid-binding protein 4, a lipid transfer protein, in both mouse and human. Our results suggest that tumor cells can receive lipid supply from omental adipocytes, and the surrounding tissues may induce tumor progression. We conclude that although omental tissue is an ideal material for reconstruction surgery, close follow-up is recommended in meningioma patients when used for cranioplasty.


Subject(s)
Meningeal Neoplasms/etiology , Meningeal Neoplasms/surgery , Meningioma/etiology , Meningioma/surgery , Omentum/pathology , Omentum/transplantation , Plastic Surgery Procedures/adverse effects , Plastic Surgery Procedures/methods , Surgical Flaps/adverse effects , Adipocytes , Aged , Animals , Cell Communication/physiology , Cell Transformation, Neoplastic , Combined Modality Therapy , Disease Models, Animal , Disease Progression , Fatty Acid-Binding Proteins/metabolism , Humans , Lung Neoplasms , Magnetic Resonance Imaging , Male , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Mice , Middle Aged , Neoplasm Recurrence, Local , Omentum/cytology , Tumor Cells, Cultured
20.
J Histochem Cytochem ; 50(4): 455-62, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897798

ABSTRACT

The thymus is a heterogeneous immune organ in which immature T-cells develop and eventually specialize to make certain immune responses of their own. Among various types of stromal cells in the thymus, thymic epithelial cells (TECs) have a crucially important function for presenting self-antigens and secreting cytokines to thymocytes for their maturation into T-cells. In this study we show that the p73 gene, a homologue of the tumor suppressor gene p53, was expressed in the nucleus of the human TEC in vivo and in TEC lines in vitro. Because p73 has the capacity to be a transactivator like p53, it may contribute to T-cell development in the context of TEC biology as regulated in the cell cycle and apoptosis.


Subject(s)
DNA-Binding Proteins/biosynthesis , Epithelial Cells/metabolism , Nuclear Proteins/biosynthesis , Thymus Gland/metabolism , Cell Line , Child, Preschool , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Thymus Gland/cytology , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL