1
|
Kuo KL, Chang SJ, Kwan AL, Chai CY. Correlation between levels of clock protein expression and effects on temozolomide-resistant glioblastoma and tumor progression. Hum Cell 2025; 38:75. [PMID: 40123038 DOI: 10.1007/s13577-025-01205-y] [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: 10/14/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Glioblastoma (GBM) is the most common malignant intracranial neoplasm. Treatment with surgical resection and concurrent chemoradiotherapy may not achieve satisfactory results in life expectancy. Temozolomide (TMZ) chemoresistance is one of the most common reasons for treatment failure, but the role of the circadian cycle and autophagic pathways in this phenomenon is unknown. This study investigated the relationship between the circadian cycle and autophagic pathways in GBM and its TMZ chemoresistance counterpart. The predictive potential of NR1D1 and MGMT was analyzed by using 631 glioma cases derived from the TCGA GBM dataset. Human GBM cell lines (U-87 MG, GBM 8401) and their TMZ chemoresistance counterparts were used for MGMT, circadian proteins (CLOCK, BMAL1, NR1D1), and LC3B analysis. In addition, immunohistochemical staining for NR1D1 was performed in 78 GBM samples, and the results were analyzed with patients' clinicopathological parameters. Results revealed a decrease in NR1D1 expression in GBM cells which could enhance TMZ chemosensitivity. Different expressions of autophagic markers were also noted in GBM cell lines with and without TMZ chemoresistance, indicating a significant role for NR1D1 in TMZ chemoresistance in the GBM cell line. In addition, higher expression of NR1D1 in tumor samples was correlated with poor prognosis and shorter survival. In conclusion, high levels of NR1D1 not only could predict poor prognosis but it could also be used as a chemosensitizer for TMZ in GBM patients.
Collapse
Affiliation(s)
- Keng-Liang Kuo
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Jyuan Chang
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| |
Collapse
|
2
|
Sun G, Cao Y, Dai X, Li M, Guo J. Hsc70 Interacts with β4GalT5 to Regulate the Growth of Gliomas. Neuromolecular Med 2019; 21:33-41. [PMID: 30607818 DOI: 10.1007/s12017-018-08520-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/15/2018] [Indexed: 12/22/2022]
Abstract
Heat shock cognate protein 70 (Hsc70) is a key mediator for the maintenance of intracellular proteins and regulates cellular activities. And it is elevated in various tumor tissues including glioma, which is closely related to the malignancy and poor prognosis of the tumors. However, the effects of Hsc70 on gliomas and its regulatory mechanism have not yet been elucidated. In the present study, we found that Hsc70 was overexpressed in glioma tissues and cultured glioma cells. Furthermore, Hsc70 expression exhibited positive correlation with the grades of gliomas. Knockdown of Hsc70 could effectively inhibit cell proliferation and increase cell apoptosis. Furthermore, we identified that β4GalT5 was a critical target for Hsc70-mediated anti-glioma effects. Blocking β4GalT5 activity could effectively reverse the anti-tumor effect of Hsc70. Taken together, these data indicate that Hsc70 regulates β4GalT5 levels, and possibly plays a role in cell proliferation and apoptosis of glioma.
Collapse
Affiliation(s)
- Guan Sun
- Department of Neurosurgery, Yancheng City No. 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224001, People's Republic of China
| | - Ying Cao
- Department of Ear-Nose-Throat, The Second People's Hospital of Huai'An, Huai'An Affiliated Hospital of Xuzhou Medical University, Huai'an, People's Republic of China
| | - Xueliang Dai
- Department of Neurosurgery, Zoucheng Peoples' Hospital, Zoucheng, People's Republic of China
| | - Min Li
- Department of Neurosurgery, Jiangning Hospital Affiliated with Nanjing Medical University, Nanjing, 211100, People's Republic of China.
| | - Jun Guo
- Department of Neurosurgery, Yancheng City No. 1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224001, People's Republic of China.
| |
Collapse
|
3
|
Chen J, Shen C, Shi J, Shen J, Chen W, Sun J, Fan S, Bei Y, Xu P, Chang H, Jiang R, Hua L, Ji B, Huang Q. Knockdown of DIXDC1 Inhibits the Proliferation and Migration of Human Glioma Cells. Cell Mol Neurobiol 2017; 37:1009-1019. [PMID: 27817168 PMCID: PMC11482149 DOI: 10.1007/s10571-016-0433-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/13/2016] [Indexed: 01/19/2023]
Abstract
DIX domain containing 1 (DIXDC1), the human homolog of coiled-coil-DIX1 (Ccd1), is a positive regulator of Wnt signaling pathway. Recently, it was found to act as a candidate oncogene in colon cancer, non-small-cell lung cancer, and gastric cancer. In this study, we aimed to investigate the clinical significance of DIXDC1 expression in human glioma and its biological function in glioma cells. Western blot and immunohistochemistry analysis showed that DIXDC1 was overexpressed in glioma tissues and glioma cell lines. The expression level of DIXDC1 was evidently linked to glioma pathological grade and Ki-67 expression. Kaplan-Meier curve showed that high expression of DIXDC1 may lead to poor outcome of glioma patients. Serum starvation and refeeding assay indicated that the expression of DIXDC1 was associated with cell cycle. To determine whether DIXDC1 could regulate the proliferation and migration of glioma cells, we transfected glioma cells with interfering RNA-targeting DIXDC1; investigated cell proliferation with Cell Counting Kit (CCK)-8, flow cytometry assays, and colony formation analyses; and investigated cell migration with wound healing assays and transwell assays. According to our data, knockdown of DIXDC1 significantly inhibited proliferation and migration of glioma cells. These data implied that DIXDC1 might participate in the development of glioma, suggesting that DIXDC1 can become a potential therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Jianguo Chen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Chaoyan Shen
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jinlong Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jianhong Shen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Wenjuan Chen
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jie Sun
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Shaocheng Fan
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuanqi Bei
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Peng Xu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Hao Chang
- Department of Neurosurgery, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, Jiangsu, People's Republic of China
| | - Rui Jiang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Lu Hua
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China
| | - Bin Ji
- Department of Radiotherapy and Oncology, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Qingfeng Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Xisi Road No. 20, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Metabolomic Screening of Tumor Tissue and Serum in Glioma Patients Reveals Diagnostic and Prognostic Information. Metabolites 2015; 5:502-20. [PMID: 26389964 PMCID: PMC4588809 DOI: 10.3390/metabo5030502] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/20/2015] [Accepted: 09/06/2015] [Indexed: 01/19/2023] Open
Abstract
Glioma grading and classification, today based on histological features, is not always easy to interpret and diagnosis partly relies on the personal experience of the neuropathologists. The most important feature of the classification is the aimed correlation between tumor grade and prognosis. However, in the clinical reality, large variations exist in the survival of patients concerning both glioblastomas and low-grade gliomas. Thus, there is a need for biomarkers for a more reliable classification of glioma tumors as well as for prognosis. We analyzed relative metabolite concentrations in serum samples from 96 fasting glioma patients and 81 corresponding tumor samples with different diagnosis (glioblastoma, oligodendroglioma) and grade (World Health Organization (WHO) grade II, III and IV) using gas chromatography-time of flight mass spectrometry (GC-TOFMS). The acquired data was analyzed and evaluated by pattern recognition based on chemometric bioinformatics tools. We detected feature patterns in the metabolomics data in both tumor and serum that distinguished glioblastomas from oligodendrogliomas (p(tumor) = 2.46 × 10(-8), p(serum) = 1.3 × 10(-5)) and oligodendroglioma grade II from oligodendroglioma grade III (p(tumor) = 0.01, p(serum) = 0.0008). Interestingly, we also found patterns in both tumor and serum with individual metabolite features that were both elevated and decreased in patients that lived long after being diagnosed with glioblastoma compared to those who died shortly after diagnosis (p(tum)(o)(r) = 0.006, p(serum) = 0.004; AUROCC(tumor) = 0.846 (0.647-1.000), AUROCC(serum) = 0.958 (0.870-1.000)). Metabolic patterns could also distinguish long and short survival in patients diagnosed with oligodendroglioma (p(tumor) = 0.01, p(serum) = 0.001; AUROCC(tumor) = 1 (1.000-1.000), AUROCC(serum) = 1 (1.000-1.000)). In summary, we found different metabolic feature patterns in tumor tissue and serum for glioma diagnosis, grade and survival, which indicates that, following further verification, metabolomic profiling of glioma tissue as well as serum may be a valuable tool in the search for latent biomarkers for future characterization of malignant glioma.
Collapse
|
5
|
Mendez G, Ozpinar A, Raskin J, Gultekin SH, Ross DA. Case comparison and literature review of glioblastoma: A tale of two tumors. Surg Neurol Int 2014; 5:121. [PMID: 25140280 PMCID: PMC4135538 DOI: 10.4103/2152-7806.138034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/11/2014] [Indexed: 11/25/2022] Open
Abstract
Background: Diagnosis of glioblastoma multiforme (GBM) includes a heterogeneous group of tumors. We describe two cases with histopathologically and molecularly similar tumors, but very different outcomes. We attempt to illustrate the need for improved prognostic markers for GBM. Case Description: Two patients with similar molecular profiles were retrospectively identified. The following markers were assessed: O6-methylguanine DNA methyltransferase (MGMT) methylation, isocitrate dehydrogenase (IDH) 1 and 2 status, epidermal growth factor receptor (EGFR) amplification, phosphatase and tensin homolog (PTEN) status, Ki-67, p53, and 1p/19q status. Each patient was assigned a Karnofsky performance score at presentation. Case 1 (62-year-old male) was a right temporal lobe glioblastoma with a molecular profile of amplified EGFR, normal PTEN, no IDH1/2 mutation, 28.7% MGMT promoter methylation, 5-20% Ki-67, 1p deletion, and 19q intact. The patient underwent resection followed by radiation therapy and 2 years of chemotherapy, and was asymptomatic and tumor free 5 years post diagnosis. Tumor eventually recurred and the patient expired 72 months after initial diagnosis. Case 2 (63-year-old male) was a right frontal white matter mass consistent with glioblastoma with a molecular profile of amplified EGFR, absent PTEN, no IDH1/2 mutation, 9.9% MGMT promoter methylation, 5-10% Ki-67, and 1p/19q status inconclusive. A radical subtotal resection was performed; however, 2 weeks later symptoms had returned. Subsequent imaging revealed a tumor larger than at diagnosis. The patient expired 3 months after initial diagnosis. Conclusion: The need for formulating more robust means to classify GBM tumor subtypes is paramount. Standard histopathologic and molecular analyses are costly and did not provide either of these patients with a realistic appraisal of their prognosis. Individualized whole genome testing similar to that being reported for medulloblastoma and other tumors may be preferable to the array of tests as currently utilized.
Collapse
Affiliation(s)
- Gustavo Mendez
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Alp Ozpinar
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey Raskin
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Sakir H Gultekin
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Donald A Ross
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA ; Section of Neurological Surgery, Operative Care Division, Portland Veterans Administration Hospital, Portland, OR, USA
| |
Collapse
|
6
|
Nicolasjilwan M, Hu Y, Yan C, Meerzaman D, Holder CA, Gutman D, Jain R, Colen R, Rubin DL, Zinn PO, Hwang SN, Raghavan P, Hammoud DA, Scarpace LM, Mikkelsen T, Chen J, Gevaert O, Buetow K, Freymann J, Kirby J, Flanders AE, Wintermark M. Addition of MR imaging features and genetic biomarkers strengthens glioblastoma survival prediction in TCGA patients. J Neuroradiol 2014; 42:212-21. [PMID: 24997477 DOI: 10.1016/j.neurad.2014.02.006] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/30/2014] [Accepted: 02/25/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE The purpose of our study was to assess whether a model combining clinical factors, MR imaging features, and genomics would better predict overall survival of patients with glioblastoma (GBM) than either individual data type. METHODS The study was conducted leveraging The Cancer Genome Atlas (TCGA) effort supported by the National Institutes of Health. Six neuroradiologists reviewed MRI images from The Cancer Imaging Archive (http://cancerimagingarchive.net) of 102 GBM patients using the VASARI scoring system. The patients' clinical and genetic data were obtained from the TCGA website (http://www.cancergenome.nih.gov/). Patient outcome was measured in terms of overall survival time. The association between different categories of biomarkers and survival was evaluated using Cox analysis. RESULTS The features that were significantly associated with survival were: (1) clinical factors: chemotherapy; (2) imaging: proportion of tumor contrast enhancement on MRI; and (3) genomics: HRAS copy number variation. The combination of these three biomarkers resulted in an incremental increase in the strength of prediction of survival, with the model that included clinical, imaging, and genetic variables having the highest predictive accuracy (area under the curve 0.679±0.068, Akaike's information criterion 566.7, P<0.001). CONCLUSION A combination of clinical factors, imaging features, and HRAS copy number variation best predicts survival of patients with GBM.
Collapse
Affiliation(s)
- Manal Nicolasjilwan
- Division of Neuroradiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Ying Hu
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD, United States
| | - Chunhua Yan
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD, United States
| | - Daoud Meerzaman
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Bethesda, MD, United States
| | - Chad A Holder
- Department of Radiology and Imaging Sciences Division of Neuroradiology, Emory University School of Medicine, Atlanta, GA, United States
| | - David Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Rajan Jain
- Departments of Radiology and Neurosurgery, Henry Ford, Detroit, MI, United States
| | - Rivka Colen
- Division of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Daniel L Rubin
- Department of Radiology and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA, United States
| | - Pascal O Zinn
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott N Hwang
- Neuroradiology Section, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Prashant Raghavan
- Division of Neuroradiology, University of Virginia Health System, Charlottesville, VA, United States
| | - Dima A Hammoud
- Radiology and Imaging Sciences, National Institutes of Health, Clinical Center, Bethesda, MD, United States
| | - Lisa M Scarpace
- Departments of Neurosurgery, Henry Ford, Detroit, MI, United States
| | - Tom Mikkelsen
- Departments of Neurosurgery, Henry Ford, Detroit, MI, United States
| | - James Chen
- Division of Neuroradiology, University of California, San Diego, CA, United States
| | - Olivier Gevaert
- Center for Cancer Systems Biology (CCSB) & Department of Radiology, Stanford University, Stanford, CA, United States
| | - Kenneth Buetow
- Arizona State University Life Science, Tempe, AZ, United States
| | | | - Justin Kirby
- SAIC-Frederick, Inc., Frederick, MD, United States
| | - Adam E Flanders
- Division of Neuroradiology, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Max Wintermark
- Division of Neuroradiology, University of Virginia Health System, Charlottesville, VA, United States; CHU de Vaudois, Department of Radiology, Lausanne, Switzerland.
| | | |
Collapse
|
7
|
Mao P, Hever-Jardine MP, Rahme GJ, Yang E, Tam J, Kodali A, Biswal B, Fadul CE, Gaur A, Israel MA, Spinella MJ. Serine/threonine kinase 17A is a novel candidate for therapeutic targeting in glioblastoma. PLoS One 2013; 8:e81803. [PMID: 24312360 PMCID: PMC3842963 DOI: 10.1371/journal.pone.0081803] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/16/2013] [Indexed: 01/01/2023] Open
Abstract
STK17A is a relatively uncharacterized member of the death-associated protein family of serine/threonine kinases which have previously been associated with cell death and apoptosis. Our prior work established that STK17A is a novel p53 target gene that is induced by a variety of DNA damaging agents in a p53-dependent manner. In this study we have uncovered an additional, unanticipated role for STK17A as a candidate promoter of cell proliferation and survival in glioblastoma (GBM). Unexpectedly, it was found that STK17A is highly overexpressed in a grade-dependent manner in gliomas compared to normal brain and other cancer cell types with the highest level of expression in GBM. Knockdown of STK17A in GBM cells results in a dramatic alteration in cell shape that is associated with decreased proliferation, clonogenicity, migration, invasion and anchorage independent colony formation. STK17A knockdown also sensitizes GBM cells to genotoxic stress. STK17A overexpression is associated with a significant survival disadvantage among patients with glioma which is independent of age, molecular phenotype, IDH1 mutation, PTEN loss, and alterations in the p53 pathway and partially independent of grade. In summary, we demonstrate that STK17A provides a proliferative and survival advantage to GBM cells and is a potential target to be exploited therapeutically in patients with glioma.
Collapse
Affiliation(s)
- Pingping Mao
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Mary P. Hever-Jardine
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Gilbert J. Rahme
- Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Eric Yang
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Janice Tam
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Anita Kodali
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Bijesh Biswal
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Camilo E. Fadul
- Neurology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Arti Gaur
- Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Mark A. Israel
- Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Michael J. Spinella
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
8
|
Faria MHG, Neves Filho EHC, Alves MKS, Burbano RMR, de Moraes Filho MO, Rabenhorst SHB. TP53 mutations in astrocytic gliomas: an association with histological grade, TP53 codon 72 polymorphism and p53 expression. APMIS 2012; 120:882-9. [PMID: 23009112 DOI: 10.1111/j.1600-0463.2012.02918.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 04/10/2012] [Indexed: 12/14/2022]
Abstract
TP53 mutations and polymorphisms have been widely related to many cancers as long as these alterations may impair its capacity to induce cell cycle arrest, DNA repair mechanisms, and apoptosis. Although TP53 alterations have been studied in astrocytic tumors, there is a lack of analysis considering specific TP53 mutations and their associations with p53 immunostainning, polymorphisms and their significance among the histological grades. Thus, we analyzed TP53 alterations in exons 2-11, including the codon 72 polymorphism, using DNA sequencing in 96 astrocytic gliomas (18 grade I, 20 grade II, 14 grade III, and 44 grade IV). Also, immunohistochemistry was assessed to evaluate the p53 protein expression. In this study, we found that the higher histological grades were statistically associated with TP53 mutations. Some of these mutations, such as TP53 P98T and TP53 G244S, seemed to be a specific marker for the higher grades, and the TP53 E286K mutation appears to be a World Health Organization grade III-IV progression marker. Also, the TP53 P98T mutation, in exon 4, is very likely to be important on the stabilization of the p53 protein, leading to its immunopositivity and it is potentially associated with the TP53 72Pro/Pro genotype.
Collapse
Affiliation(s)
- Mario H G Faria
- Department of Pathology and Forensic Medicine, Universidade Federal do Ceará, Fortaleza, Brazil
| | | | | | | | | | | |
Collapse
|
9
|
Abstract
The family of platelet-derived growth factors (PDGFs) plays a number of critical roles in normal embryonic development, cellular differentiation, and response to tissue damage. Not surprisingly, as it is a multi-faceted regulatory system, numerous pathological conditions are associated with aberrant activity of the PDGFs and their receptors. As we and others have shown, human gliomas, especially glioblastoma, express all PDGF ligands and both the two cell surface receptors, PDGFR-α and -β. The cellular distribution of these proteins in tumors indicates that glial tumor cells are stimulated via PDGF/PDGFR-α autocrine and paracrine loops, while tumor vessels are stimulated via the PDGFR-β. Here we summarize the initial discoveries on the role of PDGF and PDGF receptors in gliomas and provide a brief overview of what is known in this field.
Collapse
Affiliation(s)
- Inga Nazarenko
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Sanna-Maria Hede
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- (currently) Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, SE-751 85 Uppsala, Sweden
| | - Xiaobing He
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Anna Hedrén
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - James Thompson
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- Karolinska Healthcare Research Biobank (KHRBB), Clinical Pathology/Cytology, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Mikael S. Lindström
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- Karolinska Healthcare Research Biobank (KHRBB), Clinical Pathology/Cytology, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
10
|
|
11
|
Current world literature. Curr Opin Oncol 2011; 23:700-9. [PMID: 21993416 DOI: 10.1097/cco.0b013e32834d384a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|