25 results on '"Takashi Tsujiuchi"'
Search Results
2. Data from CD8+ T-cell–Mediated Immunoediting Influences Genomic Evolution and Immune Evasion in Murine Gliomas
- Author
-
Adam M. Sonabend, Peter Canoll, Raul Rabadan, Amy B. Heimberger, Maciej S. Lesniak, Craig Horbinski, Uttiya Basu, Gerson Rothschild, Ting Xiao, Catalina Lee-Chang, Edgar González-Buendia, Daniel Y. Zhang, Li Chen, Crismita Dmello, Angeliki Mela, Ganesh Rao, Aayushi Mahajan, Víctor A. Arrieta, Brice Laffleur, Takashi Tsujiuchi, Junfei Zhao, and Joshua R. Kane
- Abstract
Purpose:Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intratumor heterogeneity characteristic of gliomas, we hypothesized that CD8+ T cells mediate immunoediting in these tumors.Experimental Design:We developed retrovirus-induced PDGF+Pten−/− murine gliomas and evaluated glioma progression and tumor immunogenicity in the absence of CD8+ T cells by depleting this immune cell population. Furthermore, we characterized the genomic alterations present in gliomas that developed in the presence and absence of CD8+ T cells.Results:Upon transplantation, gliomas that developed in the absence of CD8+ T cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8+ T-cell depletion. In contrast, gliomas that developed under pressure from CD8+ T cells were able to fully engraft in both CD8+ T-cell–depleted mice and immunocompetent mice. Remarkably, gliomas developed in the absence of CD8+ T cells exhibited increased aneuploidy, MAPK pathway signaling, gene fusions, and macrophage/microglial infiltration, and showed a proinflammatory phenotype. MAPK activation correlated with macrophage/microglia recruitment in this model and in the human disease.Conclusions:Our studies indicate that, in these tumor models, CD8+ T cells influence glioma oncogenic pathways, tumor genotype, and immunogenicity. This suggests immunoediting of immunogenic tumor clones through their negative selection by CD8+ T cells during glioma formation.
- Published
- 2023
3. Figure S2 from CD8+ T-cell–Mediated Immunoediting Influences Genomic Evolution and Immune Evasion in Murine Gliomas
- Author
-
Adam M. Sonabend, Peter Canoll, Raul Rabadan, Amy B. Heimberger, Maciej S. Lesniak, Craig Horbinski, Uttiya Basu, Gerson Rothschild, Ting Xiao, Catalina Lee-Chang, Edgar González-Buendia, Daniel Y. Zhang, Li Chen, Crismita Dmello, Angeliki Mela, Ganesh Rao, Aayushi Mahajan, Víctor A. Arrieta, Brice Laffleur, Takashi Tsujiuchi, Junfei Zhao, and Joshua R. Kane
- Abstract
Expression of upstream activators of the MAPK pathway correlates with functional activation of the ERK and p38 cascades. (A) Correlation between Braf expression and pERK in tumors from mice treated with anti-CD8+ (n=8) and isotype control (n=8) antibodies. (B) Correlation between Ptpn11 expression and pERK in tumors from mice treated with anti-CD8 (n=8) and isotype control (n=8) antibodies. (C) Correlation between Nras expression and pERK in tumors from mice treated with anti-CD8 (n=8) and isotype control (n=8) antibodies. (D) Correlation between Traf6 expression and p-p38 in tumors from mice treated with anti-CD8+ (n=8) and isotype control (n=8) antibodies. r and p values by Pearson correlation coefficient. Points represent individual mice.
- Published
- 2023
4. Supplementary figures 1-15 from A Synthetic Cell-Penetrating Dominant-Negative ATF5 Peptide Exerts Anticancer Activity against a Broad Spectrum of Treatment-Resistant Cancers
- Author
-
Markus D. Siegelin, James M. Angelastro, Lloyd A. Greene, Peter Canoll, Jeffrey N. Bruce, Takashi Tsujiuchi, Lily Chau, Chang Shu, Basil A. Horst, and Georg Karpel-Massler
- Abstract
Suppl. fig. 1: Effect of CP-d/n-ATF5-S1 on ATF5 expression and stability Suppl. fig. 2: Effect of CP-d/n-ATF5-S1 on ASNS mRNA expression Suppl. fig. 3: Effect of CP-d/n-ATF5-S1 on cell viability in HL-60 cells Suppl. fig. 4: Pro-apoptotic activity of CP-d/n-ATF5-S1 in SF188 and GBM12 cells Suppl. fig. 5: Effect of CP-d/n-ATF5-S1 in HCT116 colorectal cancer Suppl. fig. 6: Effect of pan-caspase inhibition on CP-d/n-ATF5-S1 treatment Suppl. fig. 7: Effect of CP-d/n-ATF5-S1 on Bcl-2 family and Usp9X/Bag3 protein expression in A375 and PC3 Suppl. fig. 8: Effect of pan-caspase inhibition on CP-d/n-ATF5-S1-mediated down-regulation of Usp9X Suppl. fig. 9: Effect of Usp9X knock-down on apoptosis in LN229 Suppl. fig. 10: Isobologram for CP-d/n-ATF5-S1 and ABT263 Suppl. fig. 11: Effect of CP-d/n-ATF5-S1 and ABT263 on GBM12 cells Suppl. fig. 12: Knock-down of Mcl-1 sensitizes for ABT263-mediated apoptosis Suppl. fig. 13: Effects of CP-d/n-ATF5-S1 on U87MG xenograft model Suppl. fig. 14: Effects of CP-d/n-ATF5-S1 on PANC-1 and MDA-MB-231 xenograft model Suppl. fig. 15: Effect of CP-d/n-ATF5-S1 on organ toxicity
- Published
- 2023
5. Supplementary figure legend unmarked from A Synthetic Cell-Penetrating Dominant-Negative ATF5 Peptide Exerts Anticancer Activity against a Broad Spectrum of Treatment-Resistant Cancers
- Author
-
Markus D. Siegelin, James M. Angelastro, Lloyd A. Greene, Peter Canoll, Jeffrey N. Bruce, Takashi Tsujiuchi, Lily Chau, Chang Shu, Basil A. Horst, and Georg Karpel-Massler
- Abstract
Supplementary figure legend unmarked
- Published
- 2023
6. Supplementary materials and methods untracked from A Synthetic Cell-Penetrating Dominant-Negative ATF5 Peptide Exerts Anticancer Activity against a Broad Spectrum of Treatment-Resistant Cancers
- Author
-
Markus D. Siegelin, James M. Angelastro, Lloyd A. Greene, Peter Canoll, Jeffrey N. Bruce, Takashi Tsujiuchi, Lily Chau, Chang Shu, Basil A. Horst, and Georg Karpel-Massler
- Abstract
Supplementary materials and methods untracked
- Published
- 2023
7. Data from A Synthetic Cell-Penetrating Dominant-Negative ATF5 Peptide Exerts Anticancer Activity against a Broad Spectrum of Treatment-Resistant Cancers
- Author
-
Markus D. Siegelin, James M. Angelastro, Lloyd A. Greene, Peter Canoll, Jeffrey N. Bruce, Takashi Tsujiuchi, Lily Chau, Chang Shu, Basil A. Horst, and Georg Karpel-Massler
- Abstract
Purpose: Despite significant progress in cancer research, many tumor entities still have an unfavorable prognosis. Activating transcription factor 5 (ATF5) is upregulated in various malignancies and promotes apoptotic resistance. We evaluated the efficacy and mechanisms of the first described synthetic cell-penetrating inhibitor of ATF5 function, CP-d/n-ATF5-S1.Experimental Design: Preclinical drug testing was performed in various treatment-resistant cancer cells and in vivo xenograft models.Results: CP-d/n-ATF5-S1 reduced the transcript levels of several known direct ATF5 targets. It depleted endogenous ATF5 and induced apoptosis across a broad panel of treatment-refractory cancer cell lines, sparing non-neoplastic cells. CP-d/n-ATF5-S1 promoted tumor cell apoptotic susceptibility in part by reducing expression of the deubiquitinase Usp9X and led to diminished levels of antiapoptotic Bcl-2 family members Mcl-1 and Bcl-2. In line with this, CP-d/n-ATF5-S1 synergistically enhanced tumor cell apoptosis induced by the BH3-mimetic ABT263 and the death ligand TRAIL. In vivo, CP-d/n-ATF5-S1 attenuated tumor growth as a single compound in glioblastoma, melanoma, prostate cancer, and triple receptor–negative breast cancer xenograft models. Finally, the combination treatment of CP-d/n-ATF5-S1 and ABT263 significantly reduced tumor growth in vivo more efficiently than each reagent on its own.Conclusions: Our data support the idea that CP-d/n-ATF5-S1, administered as a single reagent or in combination with other drugs, holds promise as an innovative, safe, and efficient antineoplastic agent against treatment-resistant cancers. Clin Cancer Res; 22(18); 4698–711. ©2016 AACR.
- Published
- 2023
8. Supplementary Legend from CD8+ T-cell–Mediated Immunoediting Influences Genomic Evolution and Immune Evasion in Murine Gliomas
- Author
-
Adam M. Sonabend, Peter Canoll, Raul Rabadan, Amy B. Heimberger, Maciej S. Lesniak, Craig Horbinski, Uttiya Basu, Gerson Rothschild, Ting Xiao, Catalina Lee-Chang, Edgar González-Buendia, Daniel Y. Zhang, Li Chen, Crismita Dmello, Angeliki Mela, Ganesh Rao, Aayushi Mahajan, Víctor A. Arrieta, Brice Laffleur, Takashi Tsujiuchi, Junfei Zhao, and Joshua R. Kane
- Abstract
Supplementary Legend
- Published
- 2023
9. Rapid detection of the <scp> MYD88 L265P </scp> mutation for pre‐ and intra‐operative diagnosis of primary central nervous system lymphoma
- Author
-
Junya Yamaguchi, Fumiharu Ohka, Yotaro Kitano, Sachi Maeda, Kazuya Motomura, Kosuke Aoki, Kazuhito Takeuchi, Yuichi Nagata, Hikaru Hattori, Takashi Tsujiuchi, Ayako Motomura, Tomohide Nishikawa, Yuji Kibe, Keiko Shinjo, Yutaka Kondo, and Ryuta Saito
- Subjects
Cancer Research ,Oncology ,General Medicine - Published
- 2023
10. Newly established patient-derived organoid model of intracranial meningioma
- Author
-
Shintaro Yamazaki, Fumiharu Ohka, Yotaro Kitano, Kosuke Aoki, Yutaka Kondo, Takashi Tsujiuchi, Hiroyuki Shimizu, Alimu Adilijiang, Kuniaki Tanahashi, Yukihiro Shiraki, Toshihiko Wakabayashi, Kazuya Motomura, Atsushi Enomoto, Akira Kato, Ryuta Saito, Sachi Maeda, Atsushi Natsume, Masaki Hirano, Ayako Motomura, Keiko Shinjo, Junya Yamaguchi, and Yoshiteru Murofushi
- Subjects
Cancer Research ,Solitary fibrous tumor ,Malignant meningioma ,medicine.medical_treatment ,Forkhead Box Protein M1 ,Biology ,medicine.disease ,nervous system diseases ,Organoids ,Radiation therapy ,Meningioma ,Oncology ,Basic and Translational Investigations ,Benign Meningioma ,Meningeal Neoplasms ,otorhinolaryngologic diseases ,medicine ,Organoid ,FOXM1 ,Cancer research ,Humans ,Immunohistochemistry ,Neurology (clinical) ,neoplasms - Abstract
Background Recent comprehensive studies have revealed several molecular alterations that are frequently found in meningiomas. However, effective treatment reagents targeting specific molecular alterations have not yet been identified because of the limited number of representative research models of meningiomas. Methods We performed organoid cultures using meningioma cells and meningioma tumor tissues. Using immunohistochemistry and molecular analyses consisting of whole-exome sequencing, RNA-seq, and DNA methylation analyses, we compared the histological findings and molecular profiling of organoid models with those of parental tumors. Further, using these organoid models together with a public database of meningiomas, we explored molecular alterations, which are a potent treatment target for meningioma. Results We established 18 organoid models comprising of two malignant meningioma cells (HKBMM and IOMM-Lee), 10 benign meningiomas, four malignant meningiomas, and two solitary fibrous tumors (SFTs). The organoids exhibited consistent histological features and molecular profiles with those of the parental tumors. Using a public database, we identified that upregulated forkhead box M1 (FOXM1) was correlated with increased tumor proliferation. Overexpression of FOXM1 in benign meningioma organoids increased organoid proliferation; depletion of FOXM1 in malignant organoids decreased proliferation. Additionally, thiostrepton, a FOXM1 inhibitor combined with radiation therapy, significantly inhibited the proliferation of malignant meningioma organoid models. Conclusions An organoid model for meningioma enabled us to elucidate the tumor biology of meningioma along with potent treatment targets for meningioma.
- Published
- 2021
11. CD8+ T-cell–Mediated Immunoediting Influences Genomic Evolution and Immune Evasion in Murine Gliomas
- Author
-
Aayushi Mahajan, Junfei Zhao, Maciej S. Lesniak, Víctor A. Arrieta, Joshua Robert Kane, Li Chen, Angeliki Mela, Crismita Dmello, Craig Horbinski, Gerson Rothschild, Daniel Y Zhang, Ganesh Rao, Uttiya Basu, Takashi Tsujiuchi, Peter Canoll, Adam M. Sonabend, Raul Rabadan, Ting Xiao, Catalina Lee-Chang, Brice Laffleur, Amy B. Heimberger, and Edgar Gonzalez-Buendia
- Subjects
0301 basic medicine ,Cancer Research ,education.field_of_study ,Population ,Biology ,medicine.disease ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,Immunoediting ,Tumor progression ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,Cytotoxic T cell ,education ,CD8 - Abstract
Purpose: Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intratumor heterogeneity characteristic of gliomas, we hypothesized that CD8+ T cells mediate immunoediting in these tumors. Experimental Design: We developed retrovirus-induced PDGF+Pten−/− murine gliomas and evaluated glioma progression and tumor immunogenicity in the absence of CD8+ T cells by depleting this immune cell population. Furthermore, we characterized the genomic alterations present in gliomas that developed in the presence and absence of CD8+ T cells. Results: Upon transplantation, gliomas that developed in the absence of CD8+ T cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8+ T-cell depletion. In contrast, gliomas that developed under pressure from CD8+ T cells were able to fully engraft in both CD8+ T-cell–depleted mice and immunocompetent mice. Remarkably, gliomas developed in the absence of CD8+ T cells exhibited increased aneuploidy, MAPK pathway signaling, gene fusions, and macrophage/microglial infiltration, and showed a proinflammatory phenotype. MAPK activation correlated with macrophage/microglia recruitment in this model and in the human disease. Conclusions: Our studies indicate that, in these tumor models, CD8+ T cells influence glioma oncogenic pathways, tumor genotype, and immunogenicity. This suggests immunoediting of immunogenic tumor clones through their negative selection by CD8+ T cells during glioma formation.
- Published
- 2020
12. Surgical management of brain metastasis as a part of systematic metastases from adenoid cystic carcinoma of the external auditory canal: illustrative case
- Author
-
Shunichiro Kuramitsu, Kazuya Motomura, Yasuhiro Nakajima, Takashi Tsujiuchi, Ayako Motomura, Mamoru Matsuo, Nobuhisa Fukaya, Akinori Kageyama, Iori Kojima, Masasuke Ohno, and Ryuta Saito
- Subjects
General Medicine - Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) of the external auditory canal (EAC) is a rare tumor that accounts for approximately 5% of all EAC tumors. ACC is generally known as a slow-growing tumor, but patients often experience recurrence or distant metastasis in the long clinical course. While the major pattern of recurrence is pulmonary metastasis, brain metastasis of ACC of the EAC is rare. OBSERVATIONS The authors describe the case of a 72-year-old male who was diagnosed with ACC of the EAC. Approximately 7 years later, brain magnetic resonance imaging revealed an intra-axial homogenously enhancing mass lesion that had no direct connection with the skull base in the left frontal lobe. The patient underwent tumor resection and histopathological examination revealed a mixture of cribriform and tubular patterns. The image and pathological characteristics of the tumor were similar to those of primary ACC or ACC from other sites of origin. LESSONS While patients with ACC of the EAC often experience recurrence or distant metastasis in the long clinical course, they survive for a relatively long period of time, even though an optimal treatment has not been established. The authors therefore recommend surgical resection for brain metastasis of ACC of the EAC to improve neurological symptoms.
- Published
- 2021
13. CD8
- Author
-
Joshua R, Kane, Junfei, Zhao, Takashi, Tsujiuchi, Brice, Laffleur, Víctor A, Arrieta, Aayushi, Mahajan, Ganesh, Rao, Angeliki, Mela, Crismita, Dmello, Li, Chen, Daniel Y, Zhang, Edgar, González-Buendia, Catalina, Lee-Chang, Ting, Xiao, Gerson, Rothschild, Uttiya, Basu, Craig, Horbinski, Maciej S, Lesniak, Amy B, Heimberger, Raul, Rabadan, Peter, Canoll, and Adam M, Sonabend
- Subjects
Disease Models, Animal ,Mice ,Antigens, Neoplasm ,Brain Neoplasms ,Macrophages ,T-Lymphocytes ,Animals ,Humans ,Glioma ,Microglia ,CD8-Positive T-Lymphocytes ,Article ,Immune Evasion - Abstract
PURPOSE: Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intra-tumor heterogeneity characteristic of gliomas, we hypothesized that CD8(+) T-cells mediate immunoediting in these tumors. EXPERIMENTAL DESIGN: We developed retrovirus-induced PDGF(+)Pten(−/−) murine gliomas and evaluated glioma progression and tumor immunogenicity in the absence of CD8(+) T-cells by depleting this immune cell population. Furthermore, we characterized the genomic alterations present in gliomas that developed in the presence and absence of CD8(+) T-cells. RESULTS: Upon transplantation, gliomas that developed in the absence of CD8(+) T-cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8(+) T-cell depletion. In contrast, gliomas that developed under pressure from CD8(+) T-cells were able to fully engraft in both CD8(+) T-cell-depleted mice and immunocompetent mice. Remarkably, gliomas developed in the absence of CD8(+) T-cells exhibited increased aneuploidy, MAPK pathway signaling, gene fusions, and macrophage/microglial infiltration, and showed a proinflammatory phenotype. MAPK activation correlated with macrophage/microglia recruitment in this model and in the human disease. CONCLUSIONS: Our studies indicate that, in these tumor models, CD8(+) T-cells influence glioma oncogenic pathways, tumor genotype, and immunogenicity. This suggests immunoediting of immunogenic tumor clones through their negative selection by CD8(+) T-cells during glioma formation.
- Published
- 2019
14. CD8+ T-cell-mediated immunoediting influences genomic evolution and immune evasion in murine gliomas
- Author
-
Víctor A. Arrieta, Angeliki Mela, Brice Laffleur, Li Chen, Amy B. Heimberger, Edgar Gonzalez-Buendia, Peter Canoll, Uttiya Basu, Maciej S. Lesniak, Joshua Robert Kane, Adam M. Sonabend, Raul Rabadan, Craig Horbinski, Crismita Dmello, Catalina Lee-Chang, Daniel Y Zhang, Gerson Rothschild, Takashi Tsujiuchi, Junfei Zhao, Ganesh Rao, Ting Xiao, and Aayushi Mahajan
- Subjects
0303 health sciences ,education.field_of_study ,Population ,biochemical phenomena, metabolism, and nutrition ,Biology ,medicine.disease ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunoediting ,Tumor progression ,Immunity ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,bacteria ,Cytotoxic T cell ,education ,030304 developmental biology - Abstract
Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intra-tumor heterogeneity intrinsic to gliomas, we hypothesized that CD8+ T-cells mediate immunoediting in these tumors. We evaluated glioma progression in the absence of CD8+ T-cells by depleting this immune cell population in transgenic murine gliomas. Upon transplantation, gliomas that developed in the absence of CD8+ T-cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8+ T-cell depletion. Gliomas developed in absence of CD8+ T-cells exhibited increased chromosomal instability, MAPK signaling, gene fusions, and macrophage/microglial infiltration. MAPK activation correlated with macrophage/microglial recruitment in this model and in the human disease. Our results indicate that CD8+ T-cells mediate immunoediting during gliomagenesis, influencing the genomic stability of glioma and its microenvironment, leading to immune evasion.SignificanceImmune evasion renders cancer resistant to anti-tumoral immunity. Therapeutic intervention often fails for gliomas because of the plasticity of tumor cell variants that resist immune surveillance. Our results demonstrate a mechanism of immune evasion in gliomas that derives from CD8+ T-cells during the development and progression of this disease.
- Published
- 2019
15. Primary peripheral T-cell lymphoma, not otherwise specified, of the central nervous system in a child
- Author
-
Masazumi Fujii, Takashi Tsujiuchi, Seiichi Kato, Seiji Kojima, Hiroyuki Momota, Yoshiyuki Takahashi, and Toshihiko Wakabayashi
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Neurology ,Adolescent ,Central nervous system ,Peripheral T-cell lymphoma not otherwise specified ,Biology ,Fatal Outcome ,medicine ,Humans ,Bone Marrow Transplantation ,Gene Rearrangement ,Radiochemistry ,Brain Neoplasms ,Not Otherwise Specified ,Lymphoma, T-Cell, Peripheral ,General Medicine ,Gene rearrangement ,medicine.disease ,Peripheral T-cell lymphoma ,Lymphoma ,Genes, T-Cell Receptor ,medicine.anatomical_structure ,Oncology ,Neurology (clinical) ,Neoplasm Recurrence, Local ,Rare disease - 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.
- Published
- 2015
16. Karpel-Massler et al. Induction of synthetic lethality in IDH1-mutated gliomas through inhibition of Bcl-xL
- Author
-
Yiru Zhang, Kevin A. Roth, Elena Bianchetti, Franklin Garcia, Matei A. Banu, Chiaki Tsuge Ishida, Georg Karpel-Massler, Markus D. Siegelin, Peter Canoll, Chang Shu, Takashi Tsujiuchi, and Jeffrey N. Bruce
- Subjects
Male ,0301 basic medicine ,General Physics and Astronomy ,Apoptosis ,Synthetic lethality ,Gliomas ,lcsh:Science ,Cancer ,Sulfonamides ,Gene knockdown ,Aniline Compounds ,Multidisciplinary ,biology ,Brain Neoplasms ,Chemistry ,Glioma ,Isocitrate Dehydrogenase ,Female ,IDH1 ,Science ,bcl-X Protein ,Antineoplastic Agents ,Mice, Transgenic ,Bcl-xL ,Astrocytoma ,Mechanistic Target of Rapamycin Complex 1 ,News ,Central nervous system--Cancer ,General Biochemistry, Genetics and Molecular Biology ,Article ,Glutarates ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Bcl-2 ,neoplasms ,Intrinsic apoptosis ,glioblastoma ,AMPK ,General Chemistry ,medicine.disease ,nervous system diseases ,030104 developmental biology ,Cell culture ,Protein Biosynthesis ,Mutation ,Cancer research ,biology.protein ,Myeloid Cell Leukemia Sequence 1 Protein ,lcsh:Q ,Synthetic Lethal Mutations ,Anaplastic astrocytoma - Abstract
Certain gliomas often harbor a mutation in the activity center of IDH1 (R132H), which leads to the production of the oncometabolite 2-R-2-hydroxyglutarate (2-HG). In six model systems, including patient-derived stem cell-like glioblastoma cultures, inhibition of Bcl-xL induces significantly more apoptosis in IDH1-mutated cells than in wild-type IDH1 cells. Anaplastic astrocytoma samples with mutated IDH1 display lower levels of Mcl-1 than IDH1 wild-type tumors and specific knockdown of Mcl-1 broadly sensitizes glioblastoma cells to Bcl-xL inhibition-mediated apoptosis. Addition of 2-HG to glioblastoma cultures recapitulates the effects of the IDH mutation on intrinsic apoptosis, shuts down oxidative phosphorylation and reduces ATP levels in glioblastoma cells. 2-HG-mediated energy depletion activates AMPK (Threonine 172), blunting protein synthesis and mTOR signaling, culminating in a decline of Mcl-1. In an orthotopic glioblastoma xenograft model expressing mutated IDH1, Bcl-xL inhibition leads to long-term survival. These results demonstrate that IDH1-mutated gliomas are particularly vulnerable to Bcl-xL inhibition., Glioblastoma (GBM) cells are often characterized by the presence of the IDH1 R132H mutation and high expression of anti-apoptotic proteins. Here, the authors show that the inhibition of Bcl-xL is synthetically lethal in IDH1-mutated GBM models and that this effect is mediated by the oncometabolite, 2-HG, which reduces Mcl-1 protein levels.
- Published
- 2017
17. Papillary glioneuronal tumor with a high proliferative component and minigemistocytes in a child
- Author
-
Takashi Tsujiuchi, Hiroyuki Momota, Yoshie Shimoyama, Akiko Tatematsu, Masazumi Fujii, Masasuke Ohno, Toshihiko Wakabayashi, and Atsushi Natsume
- Subjects
Pathology ,medicine.medical_specialty ,Proliferation index ,biology ,medicine.diagnostic_test ,business.industry ,Brain tumor ,General Medicine ,Malignancy ,medicine.disease ,Pathology and Forensic Medicine ,Epilepsy ,Positron emission tomography ,Ki-67 ,biology.protein ,Medicine ,Neoplasm ,Immunohistochemistry ,Neurology (clinical) ,business - 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.
- Published
- 2014
18. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): late breaking abstracts
- Author
-
Sonja Althammer, Keith Steele, Marlon Rebelatto, Tze Heng Tan, Tobias Wiestler, Guenter Schmidt, Brandon Higgs, Xia Li, Li Shi, Xiaoping Jin, Joyce Antal, Ashok Gupta, Koustubh Ranade, Gerd Binning, Joaquim Bellmunt, Ronald de Wit, David J. Vaughn, Yves Fradet, Jae Lyun Lee, Lawrence Fong, Nicholas J. Vogelzang, Miguel A. Climent, Daniel P. Petrylak, Toni K. Choueiri, Andrea Necchi, Winald Gerritsen, Howard Gurney, David I. Quinn, Stéphane Culine, Cora N. Sternberg, Yabing Mai, Markus Puhlmann, Rodolfo F. Perini, Dean F. Bajorin, Padmanee Sharma, Margaret K. Callahan, Emiliano Calvo, Joseph W. Kim, Filipo de Braud, Patrick A. Ott, Petri Bono, Rathi N. Pillai, Michael Morse, Dung T. Le, Matthew Taylor, Pavlina Spilliopoulou, Johanna Bendell, Dirk Jaeger, Emily Chan, Scott J. Antonia, Paolo A. Ascierto, Delphine Hennicken, Marina Tschaika, Alex Azrilevich, Jonathan Rosenberg, Ofer Levy, Christopher Chan, Gady Cojocaru, Spencer Liang, Eran Ophir, Sudipto Ganguly, Amir Toporik, Maya Kotturi, Tal Fridman Kfir, Benjamin M. Murter, Kathryn Logronio, Liat Dassa, Ling Leung, Shirley Greenwald, Meir Azulay, Sandeep Kumar, Zoya Alteber, Xiaoyu Pan, Arthur Machlenkin, Yair Benita, Andrew W. Drake, Ayelet Chajut, Ran Salomon, Ilan Vankin, Einav Safyon, John Hunter, Zurit Levine, Mark White, Rom Leidner, Hyunseok Kang, Robert Haddad, Neil H. Segal, Lori J. Wirth, Robert L. Ferris, F. Stephen Hodi, Rachel E. Sanborn, Thomas F. Gajewski, William Sharfman, Dan McDonald, Shivani Srivastava, Xuemin Gu, Penny Phillips, Chaitali Passey, Tanguy Seiwert, Tsadik Habtetsion, Gang Zhou, Donastas Sakellariou-Thompson, Cara Haymaker, Caitlin Creasy, Mark Hurd, Naohiro Uraoka, Jaime Rodriguez Canales, Scott Koptez, Patrick Hwu, Anirban Maitra, Chantale Bernatchez, Scott M. Coyle, Kole T. Roybel, Levi J. Rupp, Stephen P. Santoro, Stephanie Secrest, Michael Spelman, Hanson Ho, Tina Gomes, Tiffany Tse, Chia Yung-Wu, Jack Taunton, Wendell Lim, Peter Emtage, Tarsem Moudgil, Carmen Ballesteros-Merino, Traci Hilton, Christopher Paustian, David Page, Walter Urba, Bernard Fox, Bryan Bell, Ashish Patel, Tove Olafsen, Daulet Satpayev, Michael Torgov, Filippo Marchioni, Jason Romero, Ziyue Karen Jiang, Charles Zamilpa, Jennifer S. Keppler, Alessandro Mascioni, Fang Jia, Chen-Yu Lee, Jean Gudas, Ryan J. Sullivan, Yujin Hoshida, Theodore Logan, Nikhil Khushalani, Anita Giobbie-Hurder, Kim Margolin, Joanna Roder, Rupal Bhatt, Henry Koon, Thomas Olencki, Thomas Hutson, Brendan Curti, Shauna Blackmon, James W. Mier, Igor Puzanov, Heinrich Roder, John Stewart, Asim Amin, Marc S. Ernstoff, Joseph I. Clark, Michael B. Atkins, Howard L. Kaufman, Jeffrey Sosman, Sabina Signoretti, David F. McDermott, Abraham A. Anderson, Mohammed M. Milhem, Robert H. I. Andtbacka, David Minor, Kevin S. Gorski, Daniel M. Baker, Omid Hamid, Emmanuel Akporiaye, Yoshinobu Koguchi, Kim Sutcliffe, Kristie Conder, Thomas Marron, Nina Bhardwaj, Linda Hammerich, Fiby George, Seunghee Kim-Schulze, Tibor Keler, Tom Davis, Elizabeth Crowley, Andres Salazar, Joshua Brody, Arta Monjazeb, Megan E. Daly, Jonathan Riess, Tianhong Li, William J. Murphy, Karen Kelly, Zhiwei Hu, Rulong Shen, Amanda Campbell, Elizabeth McMichael, Lianbo Yu, Bhuvaneswari Ramaswam, Cheryl A. London, Tian Xu, William Carson, Kathleen M. Kokolus, Elizabeth A. Repasky, Todd D. Schell, Joseph D. Drabick, David J. Messenheimer, Shawn Jensen, Mark Rubinstein, Kristina Andrijauskaite, Marzena Swiderska-syn, Kristin Lind, Agnes Choppin, Marina K. Roell, John Wrangle, Peter Rhode, Hing Wong, Shamim Ahmad, Mason Webb, Rasha Abu-Eid, Rajeev Shrimali, Vivek Verma, Atbin Doroodchi, Zuzana Berrong, David Yashar, Raed Samara, Mikayel Mkrtichyan, Samir Khleif, Steven Powell, Mark Gitau, Christopher Sumey, Andrew Terrell, Michele Lohr, Ryan K. Nowak, Steven McGraw, Ash Jensen, Miran Blanchard, Kathryn A. Gold, Ezra E. W. Cohen, Christie Ellison, Lora Black, John Lee, William Chad Spanos, Erik Wennerberg, Emily Schwitzer, Claire Lhuillier, Graeme Koelwyn, Rebecca Hiner, Lee Jones, Sandra Demaria, Vandeveer Amanda, John W. Greiner, Jeffrey Schlom, Michelle Bookstaver, Christopher M. Jewell, Andrew Gunderson, Brian Boulmay, Rui Li, Bradley Spieler, Kyle Happel, Zipei Feng, Christopher Dubay, Brenda Fisher, Sandra Aung, Eileen Mederos, Carlo B. Bifulco, Michael McNamara, Keith Bahjat, William Redmond, Augusto Ochoa, Hong-Ming Hu, Adi Mehta, Fridtjof Lund-Johansen, Frank Bedu-Addo, Greg Conn, Michael King, Panna Dutta, Robert Shepard, Mark Einstein, Sylvia Adams, Ena Wang, Ping Jin, Yelena Novik, Debra Morrison, Ruth Oratz, Franco M. Marincola, David Stroncek, Judith Goldberg, Silvia C. Formenti, Jérôme Galon, Bernhard Mlecnik, Florence Marliot, Fang-Shu Ou, Alessandro Lugli, Inti Zlobec, Tilman T. Rau, Iris D. Nagtegaal, Elisa Vink-Borger, Arndt Hartmann, Carol Geppert, Michael H. Roehrl, Prashant Bavi, Pamela S. Ohashi, Julia Y. Wang, Linh T. Nguyen, SeongJun Han, Heather L. MacGregor, Sara Hafezi-Bakhtiari, Bradley G. Wouters, Yutaka Kawakami, Boryana Papivanova, Mingli Xu, Tomonobu Fujita, Shoichi Hazama, Nobuaki Suzuki, Hiroaki Nagano, Kiyotaka Okuno, Kyogo Itoh, Eva Zavadova, Michal Vocka, Jan Spacek, Lubos Petruzelka, Bohuslav Konopasek, Pavel Dundr, Helena Skalova, Toshihiko Torigoe, Noriyuki Sato, Tomohisa Furuhata, Ichiro Takemasa, Marc Van den Eynde, Anne Jouret-Mourin, Jean-Pascal Machiels, Tessa Fredriksen, Lucie Lafontaine, Bénédicte Buttard, Sarah Church, Pauline Maby, Helen Angell, Mihaela Angelova, Angela Vasaturo, Gabriela Bindea, Anne Berger, Christine Lagorce, Prabhu S. Patel, Hemangini H. Vora, Birva Shah, Jayendrakumar B. Patel, Kruti N. Rajvik, Shashank J. Pandya, Shilin N. Shukla, Yili Wang, Guanjun Zhang, Giuseppe V. Masucci, Emilia K. Andersson, Fabio Grizzi, Luigi Laghi, Gerardo Botti, Fabiana Tatangelo, Paolo Delrio, Gennaro Cilberto, Franco Marincola, Daniel J. Sargent, Bernard A. Fox, Alain Algazi, Katy Tsai, Michael Rosenblum, Prachi Nandoskar, Amy Li, John Nonomura, Kathryn Takamura, Mary Dwyer, Erica Browning, Reneta Talia, Chris Twitty, Sharron Gargosky, Jean Campbell, Mai Le, Robert H. Pierce, Adil Daud, Robyn Gartrell, Douglas Marks, Edward Stack, Yan Lu, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Yichun Fu, Zoe Blake, Bret Taback, Basil Horst, Yvonne M. Saenger, Steven Leonardo, Keith Gorden, Ross B. Fulton, Kathryn Fraser, Takashi O. Kangas, Richard Walsh, Kathleen Ertelt, Jeremy Graff, Mark Uhlik, Jennifer S. Sims, Liang Lei, Takashi Tsujiuchi, Jeffrey N. Bruce, Peter Canoll, Anthony W Tolcher, Evan W Alley, Gurunadh Chichili, Jan E Canoll, Paul Moore, Ezio Bonvini, Syd Johnson, Sadhna Shankar, James Vasselli, Jon Wigginton, and John Powderly
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,medicine.medical_treatment ,Immunology ,Improved survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Immunology and Allergy ,Pharmacology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Non small cell ,business - Abstract
O1 Combinatorial CD8+ and PD-L1+ cell densities correlate with response and improved survival in non-small cell lung cancer (NSCLC) patients treated with durvalumab #### Sonja Althammer1, Keith Steele2, Marlon Rebelatto2, Tze Heng Tan1, Tobias Wiestler1, Guenter Schmidt1, Brandon Higgs2, Xia
- Published
- 2016
19. IMMU-42. CD8+ T-CELLS MEDIATE IMMUNOEDITING, AND INFLUENCE GENOTYPE, TUMOR ONCOGENIC PATHWAYS AND MICROENVIRONMENT DURING PROGRESSION OF MURINE GLIOMAS
- Author
-
Li Chen, Peter Canoll, Víctor A. Arrieta, Aayushi Mahajan, Ganesh Rao, Adam M. Sonabend, Gerson Rothschild, Craig Horbinski, Catalina Lee-Chang, Angeliki Mela, Daniel Y Zhang, Maciej S. Lesniak, Raul Rabadan, Takashi Tsujiuchi, Ting Xiao, Junfei Zhao, Crismita Dmello, Uttiya Basu, J. Robert Kane, Brice Laffleur, and Amy B. Heimberger
- Subjects
Cancer Research ,Immunology ,Cancer ,Biology ,medicine.disease ,Fusion gene ,Oncology ,Immunoediting ,Tumor progression ,Chromosome instability ,Glioma ,Genotype ,Cancer research ,medicine ,Cytotoxic T cell ,Neurology (clinical) - Abstract
Cancer immunoediting shapes tumor progression by the immunological selection of tumor cell variants that can evade immune recognition. Given the immune evasive cellular diversity of glioblastoma, we hypothesized that CD8+ T-cells mediate immunoediting in this tumor. We evaluated tumor progression in the absence of CD8+ T-cells by depleting this immune cell population in a transgenic murine glioma model. Tumors generated in the absence of CD8+ T-cells developed poorly in recipients with intact immunity, implying a more immunogenic profile. These tumors demonstrated increased chromosomal instability, gene fusions, MAPK signaling, and macrophage infiltration. These observations were stochastic, suggesting variability in the mode of tumor evolution in the absence of this immune effector. MAPK activation was correlated with macrophage recruitment in two transgenic murine models and the human disease. Our results indicate that CD8+ T-cells mediate a strong immunoediting selection in glioblastoma that protect against the hallmarks of cancer and drive immune evasion.
- Published
- 2019
20. A Synthetic Cell-Penetrating Dominant-Negative ATF5 Peptide Exerts Anticancer Activity against a Broad Spectrum of Treatment-Resistant Cancers
- Author
-
Takashi Tsujiuchi, James M. Angelastro, Lily Chau, Chang Shu, Markus D. Siegelin, Peter Canoll, Jeffrey N. Bruce, Lloyd A. Greene, Georg Karpel-Massler, and Basil A. Horst
- Subjects
0301 basic medicine ,Cancer Research ,Cell ,Activating transcription factor ,Drug Resistance ,Apoptosis ,Cell-Penetrating Peptides ,Pharmacology ,TNF-Related Apoptosis-Inducing Ligand ,Prostate cancer ,Mice ,0302 clinical medicine ,Cancer ,Membrane Potential, Mitochondrial ,Sulfonamides ,Tumor ,Aniline Compounds ,Melanoma ,Drug Synergism ,Activating Transcription Factors ,Tumor Burden ,Mitochondrial ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Caspases ,Gene Knockdown Techniques ,Development of treatments and therapeutic interventions ,Biotechnology ,Cell Survival ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Biology ,Membrane Potential ,Article ,Cell Line ,03 medical and health sciences ,Rare Diseases ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Neoplastic ,Animal ,medicine.disease ,Xenograft Model Antitumor Assays ,Brain Disorders ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Drug Resistance, Neoplasm ,Cancer cell ,Disease Models ,Cancer research ,Neoplasm ,Peptides ,Biomarkers - Abstract
Purpose: Despite significant progress in cancer research, many tumor entities still have an unfavorable prognosis. Activating transcription factor 5 (ATF5) is upregulated in various malignancies and promotes apoptotic resistance. We evaluated the efficacy and mechanisms of the first described synthetic cell-penetrating inhibitor of ATF5 function, CP-d/n-ATF5-S1. Experimental Design: Preclinical drug testing was performed in various treatment-resistant cancer cells and in vivo xenograft models. Results: CP-d/n-ATF5-S1 reduced the transcript levels of several known direct ATF5 targets. It depleted endogenous ATF5 and induced apoptosis across a broad panel of treatment-refractory cancer cell lines, sparing non-neoplastic cells. CP-d/n-ATF5-S1 promoted tumor cell apoptotic susceptibility in part by reducing expression of the deubiquitinase Usp9X and led to diminished levels of antiapoptotic Bcl-2 family members Mcl-1 and Bcl-2. In line with this, CP-d/n-ATF5-S1 synergistically enhanced tumor cell apoptosis induced by the BH3-mimetic ABT263 and the death ligand TRAIL. In vivo, CP-d/n-ATF5-S1 attenuated tumor growth as a single compound in glioblastoma, melanoma, prostate cancer, and triple receptor–negative breast cancer xenograft models. Finally, the combination treatment of CP-d/n-ATF5-S1 and ABT263 significantly reduced tumor growth in vivo more efficiently than each reagent on its own. Conclusions: Our data support the idea that CP-d/n-ATF5-S1, administered as a single reagent or in combination with other drugs, holds promise as an innovative, safe, and efficient antineoplastic agent against treatment-resistant cancers. Clin Cancer Res; 22(18); 4698–711. ©2016 AACR.
- Published
- 2016
21. Papillary glioneuronal tumor with a high proliferative component and minigemistocytes in a child
- Author
-
Hiroyuki, Momota, Masazumi, Fujii, Akiko, Tatematsu, Yoshie, Shimoyama, Takashi, Tsujiuchi, Masasuke, Ohno, Atsushi, Natsume, and Toshihiko, Wakabayashi
- Subjects
Male ,Brain Neoplasms ,Positron-Emission Tomography ,Humans ,Carbon Radioisotopes ,Astrocytoma ,Child ,Magnetic Resonance Imaging ,Neoplasms, Complex and Mixed ,Carcinoma, Papillary ,Cell Proliferation - 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.
- Published
- 2014
22. RNA Interference Therapeutics for Tumor Therapy
- Author
-
Takashi Tsujiuchi, Toshihiko Wakabayashi, Andrew D. Miller, and Atsushi Natsume
- Subjects
Messenger RNA ,T cell ,Biology ,medicine.disease ,Virology ,RNAi Therapeutics ,Genetically modified organism ,medicine.anatomical_structure ,RNA interference ,Glioma ,medicine ,Cancer research ,Nucleic acid ,Gene - Abstract
RNA interference (RNAi) was discovered as a method to control messenger RNA (mRNA) effectively in 1998, and mRNAs thereby came into the limelight as targets of gene therapies. However, there are still some problems to be worked out for mRNA targeted gene therapies, including the need to identify more effective target genes, the need to improve the introduction method of nucleic acid into target cells, and the need to further develop the administration method of preparations. In this chapter, we summarize RNAi therapeutic approaches for the treatment of solid tumors, culminating with a discussion of our O6-methylguanine–DNA methyltransferase RNAi therapeutic strategy for malignant glioma. Subsequently, we describe the perspectives toward therapy improved by using a combination of genetically modified T cell therapy and RNAi therapeutics.
- Published
- 2014
23. List of Contributors
- Author
-
Jennifer E. Adair, Jahangir Ahmed, Ghassan Alusi, Doron Amit, Scott J. Antonia, Dominick L. Auci, David A. August, Rutger K. Balvers, Lajos Baranyi, Brian C. Beard, Sebastian Brennig, Elizabeth K. Broussard, Paul D. Bryson, Andrew P. Byrnes, Denise L. Cecil, Tim Chan, Charlie Comins, Christiaan R. de Vries, Robert S. DiPaola, Clemens M.F. Dirven, Mary L. Disis, Boro Dropulic, Heather Embree, Michael W. Epperly, Juan Fueyo, Toshiyoshi Fujiwara, Hua Fung, Emmanuel Gabriel, Vidya Ganapath, Stanton L. Gerson, Steven Gill, Michal Gilon, Joe Goldufsky, Candelaria Gomez-Manzano, Jennifer Rubin Grandis, Joel S. Greenberger, John W. Greiner, Maneesh Gujrati, James L. Gulley, Amin Hajitou, Kevin J. Harrington, Arash Hatefi, Loree C. Heller, Richard Heller, Akseli Hemminki, Otto Hemminki, Ronald B. Herberman, Daniel Herendeen, Abraham Hochberg, James W. Hodge, Gregory E. Holt, Ying Huang, Insoo Hyun, Hong Jiang, Shunsuke Kagawa, Zahra Karjoo, Howard Kaufman, Chien-Chih Ke, Hans-Peter Kiem, Jonathan Kimmelman, Sarah R. Klein, Shinji Kuroda, Seong Young Kwon, Nico Lachmann, Hermann Lage, Martine L.M. Lamfers, Edmund C. Lattime, Nicholas R. Lemoine, Jonathan Lewis, John B. Liao, Steven K. Libutti, Yuan Lin, Ren-Shyan Liu, Zheng-Rong Lu, Ravi A. Madan, Imad Matouk, Alan Melcher, Andrew D. Miller, Jung-Joon Min, Thomas Moritz, James J. Mulé, Atsushi Natsume, Nastasia Nianiaris, Michael I. Nishimura, Claudia Palena, Hardev Pandha, Jessica Pastoriza, Mary C. Pinder-Schenck, Sujan Piya, Elizabeth Poplin, John A. Puskas, Thomas Quinn, Ankit Rao, Jane S. Reese, Kate Relph, Juan J. Rojas, Yvonne Saenger, Jeffrey Schlom, Kevin Shannon, Shawna A. Shirley, Guy R. Simpson, Shanthi Sivendran, Vladimir Slepushkin, Sirirurg Songsivilai, Neil Steven, Sufi Mary Thomas, Steve H. Thorne, Kwong-Yok Tsang, Takashi Tsujiuchi, Toshihiko Wakabayashi, Pin Wang, Xinxin Wang, Yaohe Wang, Robert E. Weiss, Edward White, Yohsuke Yagawa, Teerapong Yata, Ming Yuan, and Ziqiang Yuan
- Published
- 2014
24. Lack of presence of the human cytomegalovirus in human glioblastoma
- Author
-
Akira Okamoto, Yoshinori Ito, Naoaki Takemura, Tatsuya Tsurumi, Hiroki Isomura, Atsushi Natsume, Kazuya Motomura, Shinya Toyokuni, Toshihiko Wakabayashi, Yoriko Yamashita, and Takashi Tsujiuchi
- Subjects
Human cytomegalovirus ,Adult ,Male ,Adolescent ,viruses ,Cytomegalovirus ,In situ hybridization ,Biology ,medicine.disease_cause ,Real-Time Polymerase Chain Reaction ,Pathology and Forensic Medicine ,medicine ,Humans ,Child ,In Situ Hybridization, Fluorescence ,Southern blot ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,Brain Neoplasms ,Middle Aged ,medicine.disease ,Virology ,Molecular biology ,Real-time polymerase chain reaction ,Cell culture ,DNA, Viral ,Immunohistochemistry ,Female ,Carcinogenesis ,Glioblastoma ,Fluorescence in situ hybridization - Abstract
Recent reports have indicated human cytomegalovirus (HCMV) to be associated with human glioblastoma carcinogenesis. In established examples of viral carcinogenesis, viral DNA and one or more of its products have been detected in most tumor cells of biopsies in the majority of cases. To test whether HCMV is associated with human glioblastoma based on this criterion, we measured the number of viral DNA molecules per cell in both frozen and paraffin-embedded tumor biopsies from 58 patients using real-time quantitative PCR (QPCR). Immunohistochemical and fluorescence in situ hybridization (FISH) to detect HCMV proteins and genome was performed in 10 cases using formalin-fixed paraffin-embedded glioblastoma tissues. Southern blotting using DNA extracted from four glioblastoma cell lines together with immunoblotting using the four cell lines and five glioblastoma tissue samples were also performed. We further confirmed the immunoblot bands using liquid chromatography-tandem mass spectrometry assay. As a result, HCMV DNA was not detected in the tumor cells from any of the glioblastoma cases by QPCR detecting two different HCMV genes, in clear contrast to samples from patients with HCMV infection. Southern blotting and immunoblotting of cell lines and FISH using paraffin sections were all negative. However, immunoblotting and immunohistochemistry using tissue samples were partly positive, but HCMV proteins were not detected by proteomic analysis, suggesting false positivity of the analyses. As our QPCR analysis could detect 10 copies of HCMV DNA mixed with DNA extracted from 10(4) HCMV-negative cells, we conclude that HCMV is not persistent, at least in the tumor cells, of developed human glioblastoma.
- Published
- 2013
25. Induction of synthetic lethality in IDH1-mutated gliomas through inhibition of Bcl-xL.
- Author
-
Karpel-Massler, Georg, Tsuge Ishida, Chiaki, Bianchetti, Elena, Yiru Zhang, Chang Shu, Takashi Tsujiuchi, Banu, Matei A., Garcia, Franklin, Roth, Kevin A., Bruce, Jeffrey N., Canoll, Peter, and Siegelin, Markus D.
- Subjects
GLIOMAS ,MTOR protein ,OXIDATIVE phosphorylation ,PROTEIN synthesis - Abstract
Certain gliomas often harbor a mutation in the activity center of IDH1 (R132H), which leads to the production of the oncometabolite 2-R-2-hydroxyglutarate (2-HG). In six model systems, including patient-derived stem cell-like glioblastoma cultures, inhibition of Bcl-xL induces significantly more apoptosis in IDH1-mutated cells than in wild-type IDH1 cells. Anaplastic astrocytoma samples with mutated IDH1 display lower levels of Mcl-1 than IDH1 wild-type tumors and specific knockdown of Mcl-1 broadly sensitizes glioblastoma cells to Bcl-xL inhibition-mediated apoptosis. Addition of 2-HG to glioblastoma cultures recapitulates the effects of the IDH mutation on intrinsic apoptosis, shuts down oxidative phosphorylation and reduces ATP levels in glioblastoma cells. 2-HG-mediated energy depletion activates AMPK (Threonine 172), blunting protein synthesis and mTOR signaling, culminating in a decline of Mcl-1. In an orthotopic glioblastoma xenograft model expressing mutated IDH1, Bcl-xL inhibition leads to long-term survival. These results demonstrate that IDH1-mutated gliomas are particularly vulnerable to Bcl-xL inhibition. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.