1
|
Cakmak P, Jurmeister P, Divé I, Zeiner PS, Steinbach JP, Fenton TR, Plate KH, Czabanka M, Harter PN, Weber KJ. DNA methylation-based analysis reveals accelerated epigenetic aging in giant cell-enriched adult-type glioblastoma. Clin Epigenetics 2024; 16:179. [PMID: 39663543 PMCID: PMC11636044 DOI: 10.1186/s13148-024-01793-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Giant cell (gc)-enriched glioblastoma (gcGB) represents a distinct histological variant of isocitrate dehydrogenase wild-type adult-type glioblastoma with notable enlarged mono- or multinuclear tumor cells. While some studies suggest a survival advantage for gcGB patients, the underlying causes remain elusive. GcGBs are associated with TP53 mutations, and gcs were shown to accumulate DNA double-strand breaks and show deficient mitosis, potentially triggering cellular senescence programs. Epigenetic clocks have emerged as valuable tools for assessing tumor-induced age acceleration (DNAMethAgeAcc), which has lately proved itself as prognostic biomarker in glioblastoma. Our study aimed to comprehensively analyze the methylome and key metabolic proteins of gcGBs, hypothesizing that they undergo cellular aging programs compared to non-gcGBs. RESULTS A total of 310 epigenetically classified GBs, including 26 gcGBs, and nine adults with malignant gliomas allocating to pediatric high-grade glioma molecular subclasses (summarized as "pediatric GB") were included. DNAMethAgeAcc was computed by subtraction of chronological patient ages from DNA methylome-derived age estimations and its increase was associated with better survival within gcGB and non-gcGB. GcGBs were significantly more often allocated to the subgroup with increased DNAMethAgeAcc and demonstrated the highest DNAMethAgeAcc. Hypothetical senescence/aging-induced changes of the tumor microenvironment were addressed by tumor deconvolution, which was able to identify a cluster enriched for tumors with increased DNAMethAgeAcc. Key metabolic protein expression did not differ between gcGB and non-gcGB and tumor with versus without increased DNAMethAgeAcc but for elevated levels of one single mitochondrial marker, anti-mitochondrial protein MT-C02, in gcGBs. CONCLUSIONS With its sped-up epigenetic aging, gcGB presented as the epigenetic oldest GB variant in our cohort. Whereas the correlation between accelerated tumor-intrinsic epigenetic aging and cellular senescence in gcGB stays elusive, fostering epigenetic aging programs in GB might be of interest for future exploration of alternative treatment options in GB patients.
Collapse
Affiliation(s)
- Pinar Cakmak
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
| | - Philipp Jurmeister
- Ludwig Maximilians University Munich, University Hospital, Institute of Pathology, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between German Cancer Research Center (DKFZ) and University/University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Iris Divé
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Pia S Zeiner
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Department of Neurology, Frankfurt, Germany
| | - Joachim P Steinbach
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Neurooncology, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Tim R Fenton
- Somers Cancer Research, Southampton General Hospital, Southampton, UK
| | - Karl H Plate
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany
| | - Marcus Czabanka
- Goethe University Frankfurt, University Hospital, Department of Neurosurgery, Frankfurt, Germany
| | - Patrick N Harter
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between German Cancer Research Center (DKFZ) and University/University Hospital, Ludwig Maximilians University Munich, Munich, Germany
- Ludwig Maximilians University Munich, University Hospital, Center for Neuropathology and Prion Research, Munich, Germany
| | - Katharina J Weber
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt, Germany.
- Goethe University Frankfurt, Frankfurt Cancer Institute (FCI), Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt, Germany.
| |
Collapse
|
2
|
Bonnett SA, Rosenbloom AB, Ong GT, Conner M, Rininger AB, Newhouse D, New F, Phan CQ, Ilcisin S, Sato H, Lyssand JS, Geiss G, Beechem JM. Ultra High-plex Spatial Proteogenomic Investigation of Giant Cell Glioblastoma Multiforme Immune Infiltrates Reveals Distinct Protein and RNA Expression Profiles. CANCER RESEARCH COMMUNICATIONS 2023; 3:763-779. [PMID: 37377888 PMCID: PMC10155752 DOI: 10.1158/2767-9764.crc-22-0396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/20/2023] [Accepted: 04/04/2023] [Indexed: 06/29/2023]
Abstract
A deeper understanding of complex biological processes, including tumor development and immune response, requires ultra high-plex, spatial interrogation of multiple "omes". Here we present the development and implementation of a novel spatial proteogenomic (SPG) assay on the GeoMx Digital Spatial Profiler platform with next-generation sequencing readout that enables ultra high-plex digital quantitation of proteins (>100-plex) and RNA (whole transcriptome, >18,000-plex) from a single formalin-fixed paraffin-embedded (FFPE) sample. This study highlighted the high concordance, R > 0.85 and <15% change in sensitivity between the SPG assay and the single-analyte assays on various cell lines and tissues from human and mouse. Furthermore, we demonstrate that the SPG assay was reproducible across multiple users. When used in conjunction with advanced cellular neighborhood segmentation, distinct immune or tumor RNA and protein targets were spatially resolved within individual cell subpopulations in human colorectal cancer and non-small cell lung cancer. We used the SPG assay to interrogate 23 different glioblastoma multiforme (GBM) samples across four pathologies. The study revealed distinct clustering of both RNA and protein based on pathology and anatomic location. The in-depth investigation of giant cell glioblastoma multiforme (gcGBM) revealed distinct protein and RNA expression profiles compared with that of the more common GBM. More importantly, the use of spatial proteogenomics allowed simultaneous interrogation of critical protein posttranslational modifications alongside whole transcriptomic profiles within the same distinct cellular neighborhoods. Significance We describe ultra high-plex spatial proteogenomics; profiling whole transcriptome and high-plex proteomics on a single FFPE tissue section with spatial resolution. Investigation of gcGBM versus GBM revealed distinct protein and RNA expression profiles.
Collapse
Affiliation(s)
| | | | | | - Mark Conner
- NanoString Technologies, Seattle, Washington
| | | | | | - Felicia New
- NanoString Technologies, Seattle, Washington
| | - Chi Q. Phan
- NanoString Technologies, Seattle, Washington
| | | | - Hiromi Sato
- NanoString Technologies, Seattle, Washington
| | | | - Gary Geiss
- NanoString Technologies, Seattle, Washington
| | | |
Collapse
|
3
|
Aurora B Kinase Inhibition by AZD1152 Concomitant with Tumor Treating Fields Is Effective in the Treatment of Cultures from Primary and Recurrent Glioblastomas. Int J Mol Sci 2023; 24:ijms24055016. [PMID: 36902447 PMCID: PMC10003311 DOI: 10.3390/ijms24055016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Tumor Treating Fields (TTFields) were incorporated into the treatment of glioblastoma, the most malignant brain tumor, after showing an effect on progression-free and overall survival in a phase III clinical trial. The combination of TTFields and an antimitotic drug might further improve this approach. Here, we tested the combination of TTFields with AZD1152, an Aurora B kinase inhibitor, in primary cultures of newly diagnosed (ndGBM) and recurrent glioblastoma (rGBM). AZD1152 concentration was titrated for each cell line and 5-30 nM were used alone or in addition to TTFields (1.6 V/cm RMS; 200 kHz) applied for 72 h using the inovitro™ system. Cell morphological changes were visualized by conventional and confocal laser microscopy. The cytotoxic effects were determined by cell viability assays. Primary cultures of ndGBM and rGBM varied in p53 mutational status; ploidy; EGFR expression and MGMT-promoter methylation status. Nevertheless; in all primary cultures; a significant cytotoxic effect was found following TTFields treatment alone and in all but one, a significant effect after treatment with AZD1152 alone was also observed. Moreover, in all primary cultures the combined treatment had the most pronounced cytotoxic effect in parallel with morphological changes. The combined treatment of TTFields and AZD1152 led to a significant reduction in the number of ndGBM and rGBM cells compared to each treatment alone. Further evaluation of this approach, which has to be considered as a proof of concept, is warranted, before entering into early clinical trials.
Collapse
|
4
|
Xue C, Zhang B, Deng J, Liu X, Li S, Zhou J. Differentiating Giant Cell Glioblastoma from Classic Glioblastoma With Diffusion-Weighted Imaging. World Neurosurg 2020; 146:e473-e478. [PMID: 33127573 DOI: 10.1016/j.wneu.2020.10.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Differential diagnosis of giant cell glioblastoma (GC) and classic glioblastoma (GBM) using conventional radiological modalities is difficult. This study aimed to use diffusion-weighted imaging (DWI) to distinguish GC from GBM and thereby improve the accuracy of preoperative assessment of patients with GB. METHODS The clinical, magnetic resonance imaging, and pathologic data of 12 patients with GC and 21 patients with GBM were retrospectively analyzed. Independent sample t tests were used to compare the minimum apparent diffusion coefficient (ADCmin) and the normalized apparent diffusion coefficients (nADC) of the 2 tumor types. Receiver operating curve (ROC) analysis was used to assess the diagnostic efficacy of ADCmin and nADC values. RESULTS Compared with that of the classic GBM group, the ADCmin (0.98 ± 0.14 vs. 0.80 ± 0.19×10-3 mm2/second, P = 0.007) and nADC (1.42 ± 0.25 vs. 1.17 ± 0.25, P = 0.011) of the GC group were significantly higher. ROC curve analysis showed that the maximum area under the curve of ADCmin and nADC were 0.800 ± 0.080 and 0.778 ± 0.082, respectively. The sensitivity, specificity, and accuracy distinguishing GC and classic GBM was best (83.33%, 76.19%, and 78.79%, respectively) when ADCmin = 0.84×10-3 mm2/second (maximum area under the ROC, 0.800). Its positive and negative predictive values under this condition were 88.89% and 66.67%, respectively. CONCLUSIONS By distinguishing GC from classic GBM, the ADCmin parameter of DWI can improve the accuracy of the preoperative differential diagnosis of the 2 tumor types.
Collapse
Affiliation(s)
- Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
| | - Juan Deng
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
| | - Xianwang Liu
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
5
|
The therapeutic potential of Aurora kinases targeting in glioblastoma: from preclinical research to translational oncology. J Mol Med (Berl) 2020; 98:495-512. [PMID: 32219470 DOI: 10.1007/s00109-020-01895-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma is the most common aggressive primary brain tumor. Standard care includes maximal safe surgical resection, radiation, and chemotherapy with temozolomide. However, the impact of this therapeutic approach on patient survival is disappointing and poor outcomes are frequently observed. Therefore, new therapeutic targets are needed to treat this potentially deadly tumor. Aurora kinases are one of today's most sought-after classes of therapeutic targets to glioblastoma therapy. They are a family of proteins composed of three members: Aurora-A, Aurora-B, and Aurora-C that play different roles in the cell division through regulation of chromosome segregation. Deregulation of these genes has been reported in glioblastoma and a progressive number of studies have shown that inhibition of these proteins could be a promising strategy for the treatment of this tumor. This review discusses the preclinical and early clinical findings on the potential use of the Aurora kinases as new targets for the treatment of glioblastoma. KEY MESSAGES: GBM is a very aggressive tumor with limited therapeutic options. Aurora kinases are a family of serine/threonine kinases implicated in GBM pathology. Aurora kinases are critical for glioblastoma cell growth, apoptosis, and chemoresistance. Inhibition of Aurora kinases has a synergistic or sensitizing effect with chemotherapy drugs, radiotherapy, or with other targeted molecules in GBM. Several Aurora kinase inhibitors are currently in clinical trials.
Collapse
|
6
|
Delbaz A, Chen M, Jen FEC, Schulz BL, Gorse AD, Jennings MP, St John JA, Ekberg JAK. Neisseria meningitidis Induces Pathology-Associated Cellular and Molecular Changes in Trigeminal Schwann Cells. Infect Immun 2020; 88:e00955-19. [PMID: 31964742 PMCID: PMC7093114 DOI: 10.1128/iai.00955-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Neisseria meningitidis, a common cause of sepsis and bacterial meningitis, infects the meninges and central nervous system (CNS), primarily via paracellular traversal across the blood-brain barrier (BBB) or blood-cerebrospinal fluid barrier. N. meningitidis is often present asymptomatically in the nasopharynx, and the nerves extending between the nasal cavity and the brain constitute an alternative route by which the meningococci may reach the CNS. To date, the cellular mechanisms involved in nerve infection are not fully understood. Peripheral nerve glial cells are phagocytic and are capable of eliminating microorganisms, but some pathogens may be able to overcome this protection mechanism and instead infect the glia, causing cell death or pathology. Here, we show that N. meningitidis readily infects trigeminal Schwann cells (the glial cells of the trigeminal nerve) in vitro in both two-dimensional and three-dimensional cell cultures. Infection of trigeminal Schwann cells may be one mechanism by which N. meningitidis is able to invade the CNS. Infection of the cells led to multinucleation and the appearance of atypical nuclei, with the presence of horseshoe nuclei and the budding of nuclei increasing over time. Using sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics followed by bioinformatics pathway analysis, we showed that N. meningitidis induced protein alterations in the glia that were associated with altered intercellular signaling, cell-cell interactions, and cellular movement. The analysis also suggested that the alterations in protein levels were consistent with changes occurring in cancer. Thus, infection of the trigeminal nerve by N. meningitidis may have ongoing adverse effects on the biology of Schwann cells, which may lead to pathology.
Collapse
Affiliation(s)
- Ali Delbaz
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Brisbane, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Mo Chen
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Brisbane, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Freda E-C Jen
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| | - Benjamin L Schulz
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, the University of Queensland, St. Lucia, Brisbane, Australia
| | - Alain-Dominique Gorse
- QFAB Bioinformatics, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane, Australia
| | | | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Brisbane, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Brisbane, QLD, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| |
Collapse
|
7
|
Ogawa K, Kurose A, Kamataki A, Asano K, Katayama K, Kurotaki H. Giant cell glioblastoma is a distinctive subtype of glioma characterized by vulnerability to DNA damage. Brain Tumor Pathol 2020; 37:5-13. [PMID: 31655917 PMCID: PMC7028818 DOI: 10.1007/s10014-019-00355-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022]
Abstract
Giant cell glioblastoma (GC-GBM) consists of large cells with pleomorphic nuclei. As a contrast to GC-GBM, we defined monotonous small GBM (MS-GBM) as GBM that consists of small cells with monotonous small nuclei, and compared the DNA damage as well as other pathological features. GC-GBM showed minimal invasion (< 2 mm) and focal sarcomatous areas. TERTp was wild type in GC-GBM but mutant in MS-GBM. OLIG2 expression was significantly higher in MS-GBM (P < 0.01) (77% in MS-GBM and 7% in GC-GBM). GC-GBM showed significantly higher DNA double-strand breaks (DSBs) compared with MS-GBM (P < 0.01) (76% in GC-GBM and 15% in MS-GBM). Nearly, all large cells in GC-GBM underwent DSBs. Thus, significant DSBs in GC-GBM might be induced by an innate lesser stemness characteristic and be followed by mitotic slippage, resulting in polyploidization and the large pleomorphic nuclei. We conclude that GC-GBM is a distinctive subtype of glioma characterized by its vulnerability to DNA damage and that wild-type TERTp and lower OLIG2 function might induce this feature. Notably, even large pleomorphic nuclei with severe DSBs demonstrated Ki67 positivity, which alerts pathologists to the interpretation of Ki67 positivity, because cells with large nuclei undergoing severe DSBs cannot be recognized as proliferating cells that contribute to tumor aggressiveness.
Collapse
Affiliation(s)
- Kaoru Ogawa
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 5 Zaifu, Hirosaki, 036-8562, Japan
| | - Akira Kurose
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 5 Zaifu, Hirosaki, 036-8562, Japan.
| | - Akihisa Kamataki
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 5 Zaifu, Hirosaki, 036-8562, Japan
| | - Kenichiro Asano
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kosuke Katayama
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hidekachi Kurotaki
- Department of Pathology, Aomori Prefectural Central Hospital, Aomori, Japan
| |
Collapse
|
8
|
Zhao Z, Jin G, Yao K, Liu K, Liu F, Chen H, Wang K, Gorja DR, Reddy K, Bode AM, Guo Z, Dong Z. Aurora B kinase as a novel molecular target for inhibition the growth of osteosarcoma. Mol Carcinog 2019; 58:1056-1067. [PMID: 30790360 DOI: 10.1002/mc.22993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 01/07/2023]
Abstract
Osteosarcoma is the primary human malignant tumor affecting bone. This cancer most frequently arises in children and adolescents, with a second peak in those over the age of 50. Currently, surgery followed by radiotherapy and chemotherapy are the main treatments, but long-term positive effects are very poor. Aurora B kinase is a serine/threonine kinase that is a key regulator of cell cycle and mitosis. Tissue array analysis revealed that Aurora B kinase is overexpressed in osteosarcoma compared with normal bone tissue. We developed a compound, HOI-07 (i.e., (E)-3-((E)-4-(benzo[d] [1,3]dioxol-5-yl)-2-oxobut-3-en-1-ylidene)indolin-2-one), as a specific Aurora B kinase inhibitor and examined its effectiveness against osteosarcoma cell growth in this study. This compound inhibited Aurora B kinase activity in osteosarcoma and induced apoptosis, caused G2-M phase arrest, and attenuated osteosarcoma anchorage-independent cell growth. Moreover, knocking down the expression of Aurora B effectively reduced the sensitivity of osteosarcoma to HOI-07. Results of a xenograft mouse study indicated that HOI-07 treatment effectively suppressed the growth of 143B and KHOS xenografts, without affecting the body weight of mice. The expression of phosphorylated histone H3 (Ser10) was reduced in mice treated with HOI-07. Overall, we identified HOI-07 as a specific Aurora B kinase inhibitor for osteosarcoma treatment and this compound warrants further investigation.
Collapse
Affiliation(s)
- Zhenjiang Zhao
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Guoguo Jin
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Ke Yao
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Kangdong Liu
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Fangfang Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Keke Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Dhilli Rao Gorja
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| | - Kanamata Reddy
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Zhiping Guo
- Henan Provincial Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
9
|
Conde M, Michen S, Wiedemuth R, Klink B, Schröck E, Schackert G, Temme A. Chromosomal instability induced by increased BIRC5/Survivin levels affects tumorigenicity of glioma cells. BMC Cancer 2017; 17:889. [PMID: 29282022 PMCID: PMC5745881 DOI: 10.1186/s12885-017-3932-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/18/2017] [Indexed: 01/02/2023] Open
Abstract
Background Survivin, belonging to the inhibitor of apoptosis (IAP) gene family, is abundantly expressed in tumors. It has been hypothesized that Survivin facilitates carcinogenesis by inhibition of apoptosis resulting in improved survival of tumorigenic progeny. Additionally, Survivin plays an essential role during mitosis. Together with its molecular partners Aurora B, Borealin and inner centromere protein it secures bipolar chromosome segregation. However, whether increased Survivin levels contribute to progression of tumors by inducing chromosomal instability remains unclear. Methods We overexpressed Survivin in U251-MG, SVGp12, U87-MG, HCT116 and p53-deficient U87-MGshp53 and HCT116p53−/− cells. The resulting phenotype was investigated by FACS-assisted cell cycle analysis, Western Blot analysis, confocal laser scan microscopy, proliferation assays, spectral karyotyping and in a U251-MG xenograft model using immune-deficient mice. Results Overexpression of Survivin affected cells with knockdown of p53, cells harboring mutant p53 and SV40 large T antigen, respectively, resulting in the increase of cell fractions harboring 4n and >4n DNA contents. Increased γH2AX levels, indicative of DNA damage were monitored in all Survivin-transduced cell lines, but only in p53 wild type cells this was accompanied by an attenuated S-phase entry and activation of p21waf/cip. Overexpression of Survivin caused a DNA damage response characterized by increased appearance pDNA-PKcs foci in cell nuclei and elevated levels of pATM S1981 and pCHK2 T68. Additionally, evolving structural chromosomal aberrations in U251-MG cells transduced with Survivin indicated a DNA-repair by non-homologous end joining recombination. Subcutaneous transplantation of U251-MG cells overexpressing Survivin and mycN instead of mycN oncogene alone generated tumors with shortened latency and decreased apoptosis. Subsequent SKY-analysis of Survivin/mycN-tumors revealed an increase in structural chromosomal aberrations in cells when compared to mycN-tumors. Conclusions Our data suggest that increased Survivin levels promote adaptive evolution of tumors through combining induction of genetic heterogeneity with inhibition of apoptosis. Electronic supplementary material The online version of this article (10.1186/s12885-017-3932-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marina Conde
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Susanne Michen
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ralf Wiedemuth
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Evelin Schröck
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Gabriele Schackert
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,German Cancer Consortium (DKTK), partner site Dresden; German Cancer Research Center (DKFZ), Heidelberg, Germany. .,National Center for Tumor Diseases (NCT), Dresden, Germany.
| |
Collapse
|
10
|
Pisapia DJ, Magge R, Ramakrishna R. Improved Pathologic Diagnosis-Forecasting the Future in Glioblastoma. Front Neurol 2017; 8:707. [PMID: 29312129 PMCID: PMC5742147 DOI: 10.3389/fneur.2017.00707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/07/2017] [Indexed: 01/17/2023] Open
Affiliation(s)
- David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
11
|
Hinrichs CN, Ingargiola M, Käubler T, Löck S, Temme A, Köhn-Luque A, Deutsch A, Vovk O, Stasyk O, Kunz-Schughart LA. Arginine Deprivation Therapy: Putative Strategy to Eradicate Glioblastoma Cells by Radiosensitization. Mol Cancer Ther 2017; 17:393-406. [DOI: 10.1158/1535-7163.mct-16-0807] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/08/2017] [Accepted: 07/26/2017] [Indexed: 11/16/2022]
|
12
|
APIO-EE-9 is a novel Aurora A and B antagonist that suppresses esophageal cancer growth in a PDX mouse model. Oncotarget 2017; 8:53387-53404. [PMID: 28881819 PMCID: PMC5581118 DOI: 10.18632/oncotarget.18508] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/10/2017] [Indexed: 12/14/2022] Open
Abstract
Esophageal cancer (EC) is one of the most aggressive malignancies of the upper aerodigestive tract. Over the past three decades, with advances in surgical techniques and treatment, the prognosis of esophageal cancer has only slowly improved. Thus identifying novel molecular targets and developing therapeutic agents are critical. Aurora kinases play a crucial role in mitosis and selective inhibitors might provide an effective therapeutic treatment for cancer. However, the role of Aurora kinases in EC is still inadequately studied. Here, we identified a novel compound, referred to as APIO-EE-9, which inhibits growth and colony formation and induces apoptosis of esophageal cancer cells. Using computer modeling, we found that APIO-EE-9 interacted with both Aurora A and B in the ATP-binding pocket. APIO-EE-9 inhibited both Aurora A and B kinase activities in a dose-dependent manner. Treatment with APIO-EE-9 substantially reduced the downstream Aurora kinase phosphorylation of histone H3 (Ser10), resulting in formation of multiple nuclei and centrosomes. Additionally, esophageal cancer cells expressing shAurora A or shAurora B kinase exhibited a dramatic reduction in proliferation and colony formation. Injection of these cells as xenografts in mice reduced tumor formation compared to wildtype cells. Importantly, APIO-EE-9 significantly decreased the size of esophageal patient-derived xenograft (PDX) tumors implanted in SCID mice. These results demonstrated that APIO-EE-9 is a specific Aurora kinase inhibitor that could be developed as a therapeutic agent against esophageal cancer.
Collapse
|
13
|
Galli R, Uckermann O, Temme A, Leipnitz E, Meinhardt M, Koch E, Schackert G, Steiner G, Kirsch M. Assessing the efficacy of coherent anti-Stokes Raman scattering microscopy for the detection of infiltrating glioblastoma in fresh brain samples. JOURNAL OF BIOPHOTONICS 2017; 10:404-414. [PMID: 27854107 DOI: 10.1002/jbio.201500323] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/10/2016] [Accepted: 02/21/2016] [Indexed: 05/20/2023]
Abstract
Coherent anti-Stokes Raman scattering (CARS) microscopy is an emerging technique for identification of brain tumors. However, tumor identification by CARS microscopy on bulk samples and in vivo has been so far verified retrospectively on histological sections, which only provide a gross reference for the interpretation of CARS images without matching at cellular level. Therefore, fluorescent labels were exploited for direct assessment of the interpretation of CARS images of solid and infiltrative tumors. Glioblastoma cells expressing green fluorescent protein (GFP) were used for induction of tumors in mice (n = 7). The neoplastic nature of cells imaged by CARS microscopy was unequivocally verified by addressing two-photon fluorescence of GFP on fresh brain slices and in vivo. In fresh unfixed biopsies of human glioblastoma (n = 10), the fluorescence of 5-aminolevulinic acid-induced protoporphyrin IX was used for identification of tumorous tissue. Distinctive morphological features of glioblastoma cells, i.e. larger nuclei, evident nuclear membrane and nucleolus, were identified in the CARS images of both mouse and human brain tumors. This approach demonstrates that the chemical contrast provided by CARS allows the localization of infiltrating tumor cells in fresh tissue and that the cell morphology in CARS images is useful for tumor recognition. Experimental glioblastoma expressing green fluorescent protein.
Collapse
Affiliation(s)
- Roberta Galli
- Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ortrud Uckermann
- Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74,, 01307, Dresden, Germany
| | - Achim Temme
- Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74,, 01307, Dresden, Germany
| | - Elke Leipnitz
- Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74,, 01307, Dresden, Germany
| | - Matthias Meinhardt
- Neuropathology, Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Edmund Koch
- Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Gabriele Schackert
- Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74,, 01307, Dresden, Germany
| | - Gerald Steiner
- Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- Faculty of Physics, dept. of General Physics and Spectroscopy, Vilnius University, Sauletekio av. 9 bl. 3, 10222, Vilnius, Lithuania
| | - Matthias Kirsch
- Neurosurgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74,, 01307, Dresden, Germany
- CRTD/DFG-Center for Regenerative Therapies Dresden - Cluster of Excellence, Technische Universität Dresden, Fetscherstr. 105, 01307, Dresden, Germany
| |
Collapse
|
14
|
Engineering NK Cells Modified With an EGFRvIII-specific Chimeric Antigen Receptor to Overexpress CXCR4 Improves Immunotherapy of CXCL12/SDF-1α-secreting Glioblastoma. J Immunother 2016; 38:197-210. [PMID: 25962108 DOI: 10.1097/cji.0000000000000082] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Natural killer (NK) cells are promising effector cells for adjuvant immunotherapy of cancer. So far, several preclinical studies have shown the feasibility of gene-engineered NK cells, which upon expression of chimeric antigen receptors (CARs) are redirected to otherwise NK cell-resistant tumors. Yet, we reasoned that the efficiency of an immunotherapy using CAR-modified NK cells critically relies on efficient migration to the tumor site and might be improved by the engraftment of a receptor specific for a chemokine released by the tumor. On the basis of the DNAX-activation protein 12 (DAP12), a signaling adapter molecule involved in signal transduction of activating NK cell receptors, we constructed an epidermal growth factor variant III (EGFRvIII)-CAR, designated MR1.1-DAP12 which confers specific cytotoxicity of NK cell towards EGFRvIII glioblastoma cells in vitro and to established subcutaneous U87-MG tumor xenografts. So far, infusion of NK cells with expression of MR1.1-DAP12 caused a moderate but significantly delayed tumor growth and increased median survival time when compared with NK cells transduced with an ITAM-defective CAR. Notably, the further genetic engineering of these EGFRvIII-specific NK cells with the chemokine receptor CXCR4 conferred a specific chemotaxis to CXCL12/SDF-1α secreting U87-MG glioblastoma cells. Moreover, the administration of such NK cells resulted in complete tumor remission in a number of mice and a significantly increased survival when compared with the treatment of xenografts with NK cells expressing only the EGFRvIII-specific CAR or mock control. We conclude that chemokine receptor-engineered NK cells with concomitant expression of a tumor-specific CAR are a promising tool to improve adoptive tumor immunotherapy.
Collapse
|
15
|
Hausmann C, Temme A, Cordes N, Eke I. ILKAP, ILK and PINCH1 control cell survival of p53-wildtype glioblastoma cells after irradiation. Oncotarget 2016; 6:34592-605. [PMID: 26460618 PMCID: PMC4741475 DOI: 10.18632/oncotarget.5423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/25/2015] [Indexed: 11/29/2022] Open
Abstract
The prognosis is generally poor for patients suffering from glioblastoma multiforme (GBM) due to radiation and drug resistance. Prosurvival signaling originating from focal adhesion hubs essentially contributes to therapy resistance and tumor aggressiveness. As the underlying molecular mechanisms remain largely elusive, we addressed whether targeting of the focal adhesion proteins particularly interesting new cysteine-histidine-rich 1 (PINCH1), integrin-linked kinase (ILK) and ILK associated phosphatase (ILKAP) modulates GBM cell radioresistance. Intriguingly, PINCH1, ILK and ILKAP depletion sensitized p53-wildtype, but not p53-mutant, GBM cells to radiotherapy. Concomitantly, these cells showed inactivated Glycogen synthase kinase-3β (GSK3β) and reduced proliferation. For PINCH1 and ILKAP knockdown, elevated levels of radiation-induced γH2AX/53BP1-positive foci, as a marker for DNA double strand breaks, were observed. Mechanistically, we identified radiation-induced phosphorylation of DNA protein kinase (DNAPK), an important DNA repair protein, to be dependent on ILKAP. This interaction was fundamental to radiation survival of p53-wildtype GBM cells. Conclusively, our data suggest an essential role of PINCH1, ILK and ILKAP for the radioresistance of p53-wildtype GBM cells and provide evidence for DNAPK functioning as a central mediator of ILKAP signaling. Strategies for targeting focal adhesion proteins in combination with radiotherapy might be a promising approach for patients with GBM.
Collapse
Affiliation(s)
- Christina Hausmann
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Achim Temme
- Section of Experimental Neurosurgery/Tumor Immunology, Department of Neurosurgery University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Radiation Oncology, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology, 01328 Dresden, Germany.,German Cancer Consortium (DKTK), 01307 Dresden, Germany.,German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Iris Eke
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health/National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Wiedemuth R, Klink B, Fujiwara M, Schröck E, Tatsuka M, Schackert G, Temme A. Janus face-like effects of Aurora B inhibition: antitumoral mode of action versus induction of aneuploid progeny. Carcinogenesis 2016; 37:993-1003. [PMID: 27515963 DOI: 10.1093/carcin/bgw083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 08/06/2016] [Indexed: 01/10/2023] Open
Abstract
The mitotic Aurora B kinase is overexpressed in tumors and various inhibitors for Aurora B are currently under clinical assessments. However, when considering Aurora B kinase inhibitors as anticancer drugs, their mode of action and the role of p53 status as a possible predictive factor for response still needs to be investigated. In this study, we analyzed the effects of selective Aurora B inhibition using AZD1152-HQPA/Barasertib (AZD1152) on HCT116 cells, U87-MG, corresponding isogenic p53-deficient cells and a primary glioblastoma cell line. AZD1152 treatment caused polyploidy and non-apoptotic cell death in all cell lines irrespective of p53 status and was accompanied by poly-merotelic kinetochore-microtubule attachments and DNA damage. In p53 wild-type cells a DNA damage response induced an inefficient pseudo-G1 cell cycle arrest, which was not able to halt ongoing endoreplication of cells. Of note, release of tumor cells from AZD1152 resulted in recovery of aneuploid progenies bearing numerical and structural chromosomal aberrations. Yet, AZD1152 treatment enhanced death receptor TRAIL-R2 levels in all tumor cell lines investigated. A concomitant increase of the activating natural killer (NK) cell ligand MIC A/B in p53-deficient cells and an induction of FAS/CD95 in cells containing p53 rendered AZD1152-treated cells more susceptible for NK-cell-mediated lysis. Our study mechanistically explains a p53-independent mode of action of a chemical Aurora B inhibitor and suggests a potential triggering of antitumoral immune responses, following polyploidization of tumor cells, which might constrain recovery of aneuploid tumor cells.
Collapse
Affiliation(s)
- Ralf Wiedemuth
- Department of Neurosurgery, Section of Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany, German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany and
| | - Mamoru Fujiwara
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima 772-0023, Japan
| | - Evelin Schröck
- Institute for Clinical Genetics, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany, German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany and
| | - Masaaki Tatsuka
- Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shoubara, Hiroshima 772-0023, Japan
| | - Gabriele Schackert
- Department of Neurosurgery, Section of Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany, German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany and
| | - Achim Temme
- Department of Neurosurgery, Section of Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany, German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany and
| |
Collapse
|
17
|
Oh JE, Ohta T, Nonoguchi N, Satomi K, Capper D, Pierscianek D, Sure U, Vital A, Paulus W, Mittelbronn M, Antonelli M, Kleihues P, Giangaspero F, Ohgaki H. Genetic Alterations in Gliosarcoma and Giant Cell Glioblastoma. Brain Pathol 2016; 26:517-22. [PMID: 26443480 PMCID: PMC8029477 DOI: 10.1111/bpa.12328] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
The majority of glioblastomas develop rapidly with a short clinical history (primary glioblastoma IDH wild-type), whereas secondary glioblastomas progress from diffuse astrocytoma or anaplastic astrocytoma. IDH mutations are the genetic hallmark of secondary glioblastomas. Gliosarcomas and giant cell glioblastomas are rare histological glioblastoma variants, which usually develop rapidly. We determined the genetic patterns of 36 gliosarcomas and 19 giant cell glioblastomas. IDH1 and IDH2 mutations were absent in all 36 gliosarcomas and in 18 of 19 giant cell glioblastomas analyzed, indicating that they are histological variants of primary glioblastoma. Furthermore, LOH 10q (88%) and TERT promoter mutations (83%) were frequent in gliosarcomas. Copy number profiling using the 450k methylome array in 5 gliosarcomas revealed CDKN2A homozygous deletion (3 cases), trisomy chromosome 7 (2 cases), and monosomy chromosome 10 (2 cases). Giant cell glioblastomas had LOH 10q in 50% and LOH 19q in 42% of cases. ATRX loss was detected immunohistochemically in 19% of giant cell glioblastomas, but absent in 17 gliosarcomas. These and previous results suggest that gliosarcomas are a variant of, and genetically similar to, primary glioblastomas, except for a lack of EGFR amplification, while giant cell glioblastoma occupies a hybrid position between primary and secondary glioblastomas.
Collapse
Affiliation(s)
- Ji Eun Oh
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
| | - Takashi Ohta
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
- Department of Neurological SurgeryNihon University School of MedicineTokyoJapan
| | - Naosuke Nonoguchi
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
- Department of NeurosurgeryOsaka Medical CollegeTakatsukiJapan
| | - Kaishi Satomi
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
| | - David Capper
- Department of Neuropathology, Institute of PathologyUniversity of Heidelberg, and CCU Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ)69120HeidelbergGermany
| | - Daniela Pierscianek
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
- Department of NeurosurgeryUniversity Hospital EssenEssenGermany
| | - Ulrich Sure
- Department of NeurosurgeryUniversity Hospital EssenEssenGermany
| | - Anne Vital
- Bordeaux Institute of Neuroscience, CNRS UMR 5293F‐33076BordeauxFrance
| | - Werner Paulus
- Institute of Neuropathology, University Hospital MunsterD‐48149MunsterGermany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe‐UniversityFrankfurt/MainGermany
| | - Manila Antonelli
- Department of Radiology, Oncology and Anatomic PathologySapienza University of RomeI‐00161RomeItaly
| | - Paul Kleihues
- Medical FacultyUniversity of Zurich, CH‐8091 ZurichSwitzerland
| | - Felice Giangaspero
- Department of Radiology, Oncology and Anatomic PathologySapienza University of RomeI‐00161RomeItaly
- IRCCS NeuromedI‐86077PozzilliItaly
| | - Hiroko Ohgaki
- Section of Molecular Pathology, International Agency for Research on CancerF‐69372LyonFrance
| |
Collapse
|
18
|
Soroceanu L, Matlaf L, Khan S, Akhavan A, Singer E, Bezrookove V, Decker S, Ghanny S, Hadaczek P, Bengtsson H, Ohlfest J, Luciani-Torres MG, Harkins L, Perry A, Guo H, Soteropoulos P, Cobbs CS. Cytomegalovirus Immediate-Early Proteins Promote Stemness Properties in Glioblastoma. Cancer Res 2015; 75:3065-76. [PMID: 26239477 DOI: 10.1158/0008-5472.can-14-3307] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive human brain tumor. Human cytomegalovirus (HCMV) immediate-early (IE) proteins that are endogenously expressed in GBM cells are strong viral transactivators with oncogenic properties. Here, we show how HCMV IEs are preferentially expressed in glioma stem-like cells (GSC), where they colocalize with the other GBM stemness markers, CD133, Nestin, and Sox2. In patient-derived GSCs that are endogenously infected with HCMV, attenuating IE expression by an RNAi-based strategy was sufficient to inhibit tumorsphere formation, Sox2 expression, cell-cycle progression, and cell survival. Conversely, HCMV infection of HMCV-negative GSCs elicited robust self-renewal and proliferation of cells that could be partially reversed by IE attenuation. In HCMV-positive GSCs, IE attenuation induced a molecular program characterized by enhanced expression of mesenchymal markers and proinflammatory cytokines, resembling the therapeutically resistant GBM phenotype. Mechanistically, HCMV/IE regulation of Sox2 occurred via inhibition of miR-145, a negative regulator of Sox2 protein expression. In a spontaneous mouse model of glioma, ectopic expression of the IE1 gene (UL123) specifically increased Sox2 and Nestin levels in the IE1-positive tumors, upregulating stemness and proliferation markers in vivo. Similarly, human GSCs infected with the HCMV strain Towne but not the IE1-deficient strain CR208 showed enhanced growth as tumorspheres and intracranial tumor xenografts, compared with mock-infected human GSCs. Overall, our findings offer new mechanistic insights into how HCMV/IE control stemness properties in GBM cells.
Collapse
Affiliation(s)
- Liliana Soroceanu
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California.
| | - Lisa Matlaf
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Sabeena Khan
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Armin Akhavan
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Eric Singer
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Vladimir Bezrookove
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Stacy Decker
- Department of Pediatrics and Neurosurgery, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Saleena Ghanny
- Center for Applied Genomics, Institute of Genomic Medicine, University of Medicine and Dentistry of New Jersey, Newark, New Jersey
| | - Piotr Hadaczek
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Henrik Bengtsson
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - John Ohlfest
- Department of Pediatrics and Neurosurgery, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - Maria-Gloria Luciani-Torres
- Department of Neurosciences, California Pacific Medical Center Research Institute, San Francisco, California
| | - Lualhati Harkins
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Arie Perry
- Department of Pathology, University of California, San Francisco, California
| | - Hong Guo
- Center for Applied Genomics, Institute of Genomic Medicine, University of Medicine and Dentistry of New Jersey, Newark, New Jersey
| | - Patricia Soteropoulos
- Center for Applied Genomics, Institute of Genomic Medicine, University of Medicine and Dentistry of New Jersey, Newark, New Jersey
| | - Charles S Cobbs
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Seattle, Washington.
| |
Collapse
|
19
|
Fujii S, Srivastava V, Hegde A, Kondo Y, Shen L, Hoshino K, Gonzalez Y, Wang J, Sasai K, Ma X, Katayama H, Estecio MR, Hamilton SR, Wistuba I, Issa JPJ, Sen S. Regulation of AURKC expression by CpG island methylation in human cancer cells. Tumour Biol 2015; 36:8147-8158. [PMID: 25990457 DOI: 10.1007/s13277-015-3553-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/11/2015] [Indexed: 01/11/2023] Open
Abstract
AURKC, a member of the Aurora kinase gene family, is highly expressed in testis but is either moderately expressed or repressed in most somatic cells. Varying expression of AURKC has been observed in human cancers, but the underlying mechanisms of differential expression have been investigated only to a limited extent. We investigated the role of promoter CpG methylation in the regulation of AURKC gene expression in human cancer cells, in relation to a recently reported AURKC transcription repressor PLZF/ZBTB16, implicated in transformation and tumorigenesis. AURKC and PLZF/ZBTB16 expression profiles were investigated in reference to CpG methylation status on the AURKC promoter experimentally, and also in The Cancer Genome Atlas (TCGA) dataset involving multiple cancer types. AURKC promoter showed dense to moderate hypermethylation correlating with low to moderate expression of the gene in normal somatic cells and cancer cell lines, while testis with high expression revealed marked hypo-methylation. Treatment with the demethylating agent, 5-aza-dC, but not the histone deacetylase (HDAC) inhibitor, TSA, led to elevated expression in cancer cell lines, indicating that promoter DNA methylation negatively regulates AURKC expression. High expression of PLZF in PLZF-transfected cells treated with 5-aza-dC only partially repressed expression of AURKC despite 5-aza-dC also inducing elevated PLZF expression. Analyses of the TCGA data showed differential expression of AURKC in multiple cancer types and stronger correlation of AURKC expression with CpG methylation compared to PLZF levels. These findings demonstrate that differential promoter CpG methylation is an important mechanism regulating AURKC expression in cancer cells.
Collapse
Affiliation(s)
- Satoshi Fujii
- Pathology Division, Research Center for Innovative Oncology, National Cancer Center at Kashiwa, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Vibhuti Srivastava
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA
- Molecular Genetic Technology Program, School of Health Professions, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Apurva Hegde
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA
- Program in Human and Molecular Genetics, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yutaka Kondo
- Division of Epigenomics, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681, Japan
| | - Lanlan Shen
- Department of Pediatrics, USDA Children's Nutrition Research Center-Baylor College of Medicine, 1100 Bates St., Ste. 8020, Houston, TX, 77030, USA
| | - Koyu Hoshino
- Department of Hematology and Clinical Rheumatology, Graduate School of Medical Sciences-Kumamoto University, Honjo 1-1-1, Kumamoto, 860-8556, Japan
| | - Yvette Gonzalez
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Jin Wang
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Kaori Sasai
- Department of Molecular Genetics, Okayama University Medical School, Okayama, 700-8558, Japan
| | - Xiaotu Ma
- Center for Systems Biology, Department of Molecular and Cell Biology, University of Texas at Dallas, RL10, 800 W Campbell Road, Richardson, TX, 75080, USA
| | - Hiroshi Katayama
- Department of Molecular Genetics, Okayama University Medical School, Okayama, 700-8558, Japan
| | - Marcos R Estecio
- Department of Epigenetics and Molecular Carcinogenesis, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stanley R Hamilton
- Division of Pathology and Laboratory Medicine, UT MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA
| | - Jean-Pierre J Issa
- Fels Institute for Cancer Research and Molecular Biology, Temple University, PAHB Room 154, 3307 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Subrata Sen
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Unit 2951, Life Science Plaza Building, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
20
|
Belsuzarri TAB, Araujo JFM, Catanoce AP, Neves MWF, Sola RAS, Navarro JN, Brito LG, Silva NR, Pontelli LOC, Mattos LGA, Gonçales TF, Zeviani WM, Marques RMB. Giant cells glioblastoma: case report and pathological analysis from this uncommon subtype of glioma. Rare Tumors 2015; 7:5634. [PMID: 25918606 PMCID: PMC4387352 DOI: 10.4081/rt.2015.5634] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common glial tumor of the brain system; nevertheless, the giant cell (GC) subtype is uncommon. Recent reviews report for an incidence of 1% in adults and 3% in children. The GCs usually have a better prognosis than GBM and also an increasing long-term survival rate. It is known that the diagnosis of this tumor is due to its histological findings and patterns, such as the unusual increased number of giant cells. Unfortunately, due to its rarity, the immunohistochemical and cytogenetical analysis of this tumor is not well known. Some authors also suggest that there are few subtypes of GCs and their patterns of aggressiveness could be due to cytogenetical markers. It is recognized that maximum safe resection treatment and adjuvant radiotherapy can improve survival rate (5-13 months) similar to GBM patients.
Collapse
Affiliation(s)
- Telmo A B Belsuzarri
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - João F M Araujo
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Aguinaldo P Catanoce
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Maick W F Neves
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Rodrigo A S Sola
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Juliano N Navarro
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Leandro G Brito
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Nilton R Silva
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Luis Otavio C Pontelli
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Luiz Gustavo A Mattos
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Tiago F Gonçales
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Wolnei M Zeviani
- Department of Neurosurgery, Pontifical Catholic University of Campinas , São Paulo, Brazil
| | - Renata M B Marques
- Department of Pathology, Maternity Hospital of Celso Pierro, Pontifical Catholic University of Campinas , São Paulo, Brazil
| |
Collapse
|
21
|
Wiedemuth R, Klink B, Töpfer K, Schröck E, Schackert G, Tatsuka M, Temme A. Survivin safeguards chromosome numbers and protects from aneuploidy independently from p53. Mol Cancer 2014; 13:107. [PMID: 24886358 PMCID: PMC4041913 DOI: 10.1186/1476-4598-13-107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/02/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Survivin, a member of the inhibitor of apoptosis (IAP) gene family, has a dual role in mitosis and in apoptosis. It is abundantly expressed in every human tumor, compared with normal tissues. During mitosis Survivin assembles with the chromosomal passenger complex and regulates chromosomal segregation. Here, we aim to explore whether interference with the mitotic function of Survivin is linked to p53-mediated G1 cell cycle arrest and affects chromosomal stability. METHODS In this study, we used HCT116, SBC-2, and U87-MG and generated corresponding isogenic p53-deficient cells. Retroviral vectors were used to stably knockdown Survivin. The resulting phenotype, in particular the mechanisms of cell cycle arrest and of initiation of aneuploidy, were investigated by Western Blot analysis, confocal laser scan microscopy, proliferation assays, spectral karyotyping and RNAi. RESULTS In all cell lines Survivin-RNAi did not induce instant apoptosis but caused polyplodization irrespective of p53 status. Strikingly, polyploidization after knockdown of Survivin resulted in merotelic kinetochore spindle assemblies, γH2AX-foci, and DNA damage response (DDR), which was accompanied by a transient p53-mediated G1-arrest. That p53 wild type cells specifically arrest due to DNA damage was shown by simultaneous inhibition of ATM and DNA-PK, which abolished induction of p21waf/cip. Cytogenetic analysis revealed chromosomal aberrations indicative for DNA double strand break repair by the mechanism of non-homologous end joining (NHEJ), only in Survivin-depleted cells. CONCLUSION Our findings suggest that Survivin plays an essential role in proper amphitelic kinetochore-spindle assembly and that constraining Survivin's mitotic function results in polyploidy and aneuploidy which cannot be controlled by p53. Therefore, Survivin critically safeguards chromosomal stability independently from p53.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr, 74, 01307 Dresden, Germany.
| |
Collapse
|
22
|
Oppel F, Müller N, Schackert G, Hendruschk S, Martin D, Geiger KD, Temme A. SOX2-RNAi attenuates S-phase entry and induces RhoA-dependent switch to protease-independent amoeboid migration in human glioma cells. Mol Cancer 2011; 10:137. [PMID: 22070920 PMCID: PMC3228695 DOI: 10.1186/1476-4598-10-137] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 11/09/2011] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND SOX2, a high mobility group (HMG)-box containing transcription factor, is a key regulator during development of the nervous system and a persistent marker of neural stem cells. Recent studies suggested a role of SOX2 in tumor progression. In our previous work we detected SOX2 in glioma cells and glioblastoma specimens. Herein, we aim to explore the role of SOX2 for glioma malignancy in particular its role in cell proliferation and migration. METHODS Retroviral shRNA-vectors were utilized to stably knockdown SOX2 in U343-MG and U373-MG cells. The resulting phenotype was investigated by Western blot, migration/invasion assays, RhoA G-LISA, time lapse video imaging, and orthotopic xenograft experiments. RESULTS SOX2 depletion results in pleiotropic effects including attenuated cell proliferation caused by decreased levels of cyclinD1. Also an increased TCF/LEF-signaling and concomitant decrease in Oct4 and Nestin expression was noted. Furthermore, down-regulation of focal adhesion kinase (FAK) signaling and of downstream proteins such as HEF1/NEDD9, matrix metalloproteinases pro-MMP-1 and -2 impaired invasive proteolysis-dependent migration. Yet, cells with knockdown of SOX2 switched to a RhoA-dependent amoeboid-like migration mode which could be blocked by the ROCK inhibitor Y27632 downstream of RhoA-signaling. Orthotopic xenograft experiments revealed a higher tumorigenicity of U343-MG glioma cells transduced with shRNA targeting SOX2 which was characterized by increased dissemination of glioma cells. CONCLUSION Our findings suggest that SOX2 plays a role in the maintenance of a less differentiated glioma cell phenotype. In addition, the results indicate a critical role of SOX2 in adhesion and migration of malignant gliomas.
Collapse
Affiliation(s)
- Felix Oppel
- Department of Neurosurgery, Section Experimental Neurosurgery and Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Fetscherstr 74, 01307 Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Gil-Perotin S, Haines JD, Kaur J, Marin-Husstege M, Spinetta MJ, Kim KH, Duran-Moreno M, Schallert T, Zindy F, Roussel MF, Garcia-Verdugo JM, Casaccia P. Roles of p53 and p27(Kip1) in the regulation of neurogenesis in the murine adult subventricular zone. Eur J Neurosci 2011; 34:1040-52. [PMID: 21899604 DOI: 10.1111/j.1460-9568.2011.07836.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The tumor suppressor protein p53 (Trp53) and the cell cycle inhibitor p27(Kip1) (Cdknb1) have both been implicated in regulating proliferation of adult subventricular zone (aSVZ) cells. We previously reported that genetic ablation of Trp53 (Trp53-/-) or Cdknb1 (p27(Kip1-/-) ) increased proliferation of cells in the aSVZ, but differentially affected the number of adult born neuroblasts. We therefore hypothesized that these molecules might play non-redundant roles. To test this hypothesis we generated mice lacking both genes (Trp53-/- ;p27(Kip1-/-) ) and analysed the consequences on aSVZ cells and adult neuroblasts. Proliferation and self-renewal of cultured aSVZ cells were increased in the double mutants compared with control, but the mice did not develop spontaneous brain tumors. In contrast, the number of adult-born neuroblasts in the double mutants was similar to wild-type animals and suggested a complementation of the p27(Kip1-/-) phenotype due to loss of Trp53. Cellular differences detected in the aSVZ correlated with cellular changes in the olfactory bulb and behavioral data on novel odor recognition. The exploration time for new odors was reduced in p27(Kip1-/-) mice, increased in Trp53-/- mice and normalized in the double Trp53-/- ;p27(Kip1-/-) mutants. At the molecular level, Trp53-/- aSVZ cells were characterized by higher levels of NeuroD and Math3 and by the ability to generate neurons more readily. In contrast, p27(Kip1-/-) cells generated fewer neurons, due to enhanced proteasomal degradation of pro-neural transcription factors. Together, these results suggest that p27(Kip1) and p53 function non-redundantly to modulate proliferation and self-renewal of aSVZ cells and antagonistically in regulating adult neurogenesis.
Collapse
Affiliation(s)
- Sara Gil-Perotin
- Department of Comparative Neurobiology, CIBERNED, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|