1
|
Murugan AK, Kannan S, Alzahrani AS. Immune checkpoint expression and therapeutic implications in IDH1-mutant and wild-type glioblastomas. Curr Probl Cancer 2025; 55:101182. [PMID: 39864140 DOI: 10.1016/j.currproblcancer.2025.101182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/17/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Programmed cell death protein 1 (PDCD1) and cluster of differentiation 274 (CD274) expression is implicated in escaping tumors from immune surveillance. Immune checkpoint inhibitors show promise in cancer therapy, yet their efficacy in glioblastomas, particularly with IDH1 mutations, remains unclear. This study analyzed two independent NGS datasets (n = 577 and n = 153) from TCGA to investigate the expression of PDCD1 and CD274 in glioblastomas and their relationship with IDH1 mutations. We used cBioPortal for mutation analysis, RNA seq for expression analysis, miRDB and miRabel for differential expression of miRNAs, and Kaplan-Meier for survival prediction. We found that 5.4% of glioblastomas harbored IDH1 mutations, correlating with improved overall survival (OS) (p = 2.196e-3). Different glioblastoma cohorts showed a diverse IDH1 mutational prevalence (4-31%). Despite this, IDH1Mu was consistently associated with better OS (p = 8.235e-5). Notably, PDCD1 and CD274 were statistically significantly highly expressed in both IDH1Wt (p < 0.0001) and IDH1Mu tumors (p < 0.0001), with higher expression linked to poorer survival outcomes (PDCD1: p = 0.009; CD274: p = 0.02). Differential co-expression analyses revealed distinct gene and miRNA profiles for IDH1Wt and IDH1Mu glioblastomas, with specific upregulation of PTEN and downregulation of MUC16 in IDH1Wt, and upregulation of PIK3R1 in IDH1Mu. Additionally, PIK3R1 and ITGB2 emerged as critical druggable targets. Our findings indicate that PDCD1 and CD274 are highly expressed irrespective of IDH1 mutation statuses, suggesting that glioblastomas could benefit from immunotherapy. Moreover, IDH1Mu glioblastomas may require a combination of PI3K/AKT/mTOR inhibitors and immunotherapy due to PIK3R1 overexpression.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211 Saudi Arabia
| |
Collapse
|
2
|
Fan S, Wu N, Chang S, Chen L, Sun X. The immune regulation of BCL3 in glioblastoma with mutated IDH1. Aging (Albany NY) 2022; 14:3856-3873. [PMID: 35488886 PMCID: PMC9134951 DOI: 10.18632/aging.204048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022]
Abstract
Background: Glioblastoma in the brain is the most malignant solid tumor with a poor prognosis. Screening critical targets and exploring underlying mechanisms will be a benefit for diagnoses and treatment. IDH1 mutation (R132) was used to distinguish glioblastoma grade and predict prognosis as a significant marker. However, the manner of IDH1 mutation regulating glioblastoma development was still unclear. Methods: To study the function of IDH1 mutation, multi-type sequencing data (transcriptome, methylation and copy number variation) from the GEO and TCGA database were analyzed using bioinformatics techniques. The biological functions of IDH1 mutation (R132) would be comprehensively evaluated from the regulatory networks, tumor immune microenvironment and clinical relevance. Then the analysis result would be validated by experimental techniques. Results: Compared with adjacent tissues, IDH1 was up-regulated in glioblastoma, which also positively correlated with the malignant degree and a poor prognosis. To further study the mechanism of mutated IDH1 (R132) function, 5 correlated genes (FABP5, C1RL, MIR155HG, CSTA and BCL3) were identified by different expression gene screening, enrichment analysis and network construction successively. Among them, the BCL3 was a transcription factor that may induce IDH1expression. Through calculating the correlation coefficient, it was found that in IDH1mut glioblastoma, the dendritic cell infiltration was reduced which may result in a better prognosis. In addition, the level of IDH1, FABP5, C1RL, MIR155HG, CSTA and BCL3 might also influence lymphocytes infiltration (eg. CD4+ T cell) and chemokine expression (CXCL family). Conclusions: IDH1 may participate in pathological mechanisms of glioblastoma via expression alteration or gene mutation. Furthermore, IDH1 mutation might improve prognosis via suppressing the expression of FABP5, C1RL, MIR155HG, CSTA and BCL3. Meanwhile, it was identified that BCL3 might perform similar immunomodulatory functions with IDH1 as an upstream transcript factor.
Collapse
Affiliation(s)
- Shibing Fan
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China.,Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Na Wu
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China.,Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Shichuan Chang
- Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Long Chen
- Chongqing University, Shapingba, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Walker JM, Kazempour Dehkordi S, Fracassi A, Vanschoiack A, Pavenko A, Taglialatela G, Woltjer R, Richardson TE, Zare H, Orr ME. Differential protein expression in the hippocampi of resilient individuals identified by digital spatial profiling. Acta Neuropathol Commun 2022; 10:23. [PMID: 35164877 PMCID: PMC8842950 DOI: 10.1186/s40478-022-01324-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Clinical symptoms correlate with underlying neurodegenerative changes in the vast majority of people. However, an intriguing group of individuals demonstrate neuropathologic changes consistent with Alzheimer disease (AD) yet remain cognitively normal (termed "resilient"). Previous studies have reported less overall neuronal loss, less gliosis, and fewer comorbidities in these individuals. Herein, NanoString GeoMx™ Digital Spatial Profiler (DSP) technology was utilized to investigate protein expression differences comparing individuals with dementia and AD neuropathologic change to resilient individuals. DSP allows for spatial analysis of protein expression in multiple regions of interest (ROIs) on formalin-fixed paraffin-embedded sections. ROIs in this analysis were hippocampal neurofibrillary tangle (NFT)-bearing neurons, non-NFT-bearing neurons, and their immediate neuronal microenvironments. Analyses of 86 proteins associated with CNS cell-typing or known neurodegenerative changes in 168 ROIs from 14 individuals identified 11 proteins displaying differential expression in NFT-bearing neurons of the resilient when compared to the demented (including APP, IDH1, CD68, GFAP, SYP and Histone H3). In addition, IDH1, CD68, and SYP were differentially expressed in the environment of NFT-bearing neurons when comparing resilient to demented. IDH1 (which is upregulated under energetic and oxidative stress) and PINK1 (which is upregulated in response to mitochondrial dysfunction and oxidative stress) both displayed lower expression in the environment of NFT-bearing neurons in the resilient. Therefore, the resilient display less evidence of energetic and oxidative stress. Synaptophysin (SYP) was increased in the resilient, which likely indicates better maintenance of synapses and synaptic connections. Furthermore, neurofilament light chain (NEFL) and ubiquitin c-terminal hydrolase (Park5) were higher in the resilient in the environment of NFTs. These differences all suggest healthier intact axons, dendrites and synapses in the resilient. In conclusion, resilient individuals display protein expression patterns suggestive of an environment containing less energetic and oxidative stress, which in turn results in maintenance of neurons and their synaptic connections.
Collapse
Affiliation(s)
- Jamie M. Walker
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
| | - Shiva Kazempour Dehkordi
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB, Galveston, TX USA
| | | | | | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB, Galveston, TX USA
| | - Randall Woltjer
- Department of Pathology & Laboratory Medicine, Oregon Health and Science University, Portland, OR USA
| | - Timothy E. Richardson
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
| | - Habil Zare
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Miranda E. Orr
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Salisbury VA Medical Center, Salisbury, NC 28144 USA
- Department of Internal Medicine, Wake Forest School of Medicine, 575 Patterson Ave, Winston-Salem, NC 27101 USA
| |
Collapse
|
4
|
Mazurek M, Szczepanek D, Orzyłowska A, Rola R. Analysis of Factors Affecting 5-ALA Fluorescence Intensity in Visualizing Glial Tumor Cells-Literature Review. Int J Mol Sci 2022; 23:ijms23020926. [PMID: 35055109 PMCID: PMC8779265 DOI: 10.3390/ijms23020926] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Glial tumors are one of the most common lesions of the central nervous system. Despite the implementation of appropriate treatment, the prognosis is not successful. As shown in the literature, maximal tumor resection is a key element in improving therapeutic outcome. One of the methods to achieve it is the use of fluorescent intraoperative navigation with 5-aminolevulinic acid. Unfortunately, often the level of fluorescence emitted is not satisfactory, resulting in difficulties in the course of surgery. This article summarizes currently available knowledge regarding differences in the level of emitted fluorescence. It may depend on both the histological type and the genetic profile of the tumor, which is reflected in the activity and expression of enzymes involved in the intracellular metabolism of fluorescent dyes, such as PBGD, FECH, UROS, and ALAS. The transport of 5-aminolevulinic acid and its metabolites across the blood–brain barrier and cell membranes mediated by transporters, such as ABCB6 and ABCG2, is also important. Accompanying therapies, such as antiepileptic drugs or steroids, also have an impact on light emission by tumor cells. Accurate determination of the factors influencing the fluorescence of 5-aminolevulinic acid-treated cells may contribute to the improvement of fluorescence navigation in patients with highly malignant gliomas.
Collapse
|
5
|
Molenaar RJ, Wilmink JW. IDH1/2 Mutations in Cancer Stem Cells and Their Implications for Differentiation Therapy. J Histochem Cytochem 2021; 70:83-97. [PMID: 34967233 PMCID: PMC8721574 DOI: 10.1369/00221554211062499] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH1/2) are enzymes recurrently mutated in various types of cancer, including glioma, cholangiocarcinoma, chondrosarcoma, and acute myeloid leukemia. Mutant IDH1/2 induce a block in differentiation and thereby contribute to the stemness and oncogenesis of their cells of origin. Recently, small-molecule inhibitors of mutant IDH1/2 have been Food and Drug Administration-approved for the treatment of IDH1/2-mutated acute myeloid leukemia. These inhibitors decrease the stemness of the targeted IDH1/2-mutated cancer cells and induce their differentiation to more mature cells. In this review, we elucidate the mechanisms by which mutant IDH1/2 induce a block in differentiation and the biological and clinical effects of the release into differentiation by mutant-IDH1/2 inhibitors. (J Histochem Cytochem 70:83-97, 2022).
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
6
|
van Noorden CJ, Hira VV, van Dijck AJ, Novak M, Breznik B, Molenaar RJ. Energy Metabolism in IDH1 Wild-Type and IDH1-Mutated Glioblastoma Stem Cells: A Novel Target for Therapy? Cells 2021; 10:cells10030705. [PMID: 33810170 PMCID: PMC8005124 DOI: 10.3390/cells10030705] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a redox disease. Low levels of reactive oxygen species (ROS) are beneficial for cells and have anti-cancer effects. ROS are produced in the mitochondria during ATP production by oxidative phosphorylation (OXPHOS). In the present review, we describe ATP production in primary brain tumors, glioblastoma, in relation to ROS production. Differentiated glioblastoma cells mainly use glycolysis for ATP production (aerobic glycolysis) without ROS production, whereas glioblastoma stem cells (GSCs) in hypoxic periarteriolar niches use OXPHOS for ATP and ROS production, which is modest because of the hypoxia and quiescence of GSCs. In a significant proportion of glioblastoma, isocitrate dehydrogenase 1 (IDH1) is mutated, causing metabolic rewiring, and all cancer cells use OXPHOS for ATP and ROS production. Systemic therapeutic inhibition of glycolysis is not an option as clinical trials have shown ineffectiveness or unwanted side effects. We argue that systemic therapeutic inhibition of OXPHOS is not an option either because the anti-cancer effects of ROS production in healthy cells is inhibited as well. Therefore, we advocate to remove GSCs out of their hypoxic niches by the inhibition of their binding to niches to enable their differentiation and thus increase their sensitivity to radiotherapy and/or chemotherapy.
Collapse
Affiliation(s)
- Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
- Department of Medical Biology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: ; Tel.: +31-638-639-561
| | - Vashendriya V.V. Hira
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Amber J. van Dijck
- Department of Medical Biology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Remco J. Molenaar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
- Department of Medical Oncology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
7
|
Zhong J, Kang Q, Cao Y, He B, Zhao P, Gou Y, Luo Y, He TC, Fan J. BMP4 augments the survival of hepatocellular carcinoma (HCC) cells under hypoxia and hypoglycemia conditions by promoting the glycolysis pathway. Am J Cancer Res 2021; 11:793-811. [PMID: 33791154 PMCID: PMC7994163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/16/2020] [Indexed: 04/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide although its pathogenic mechanism remains to be fully understood. Unlike normal cells, most cancer cells rely on aerobic glycolysis and are more adaptable to the microenvironment of hypoxia and hypoglycemia. Bone Morphogenetic Protein 4 (BMP4) plays important roles in regulating proliferation, differentiation, invasion and migration of HCC cells. We have recently shown that BMP4 plays an important role in regulating glucose metabolism although the effect of BMP4 on glucose metabolic reprogramming of HCC is poorly understood. In this study, we found that BMP4 was highly expressed in HCC tumor tissues, as well as HCC cell lines that were tolerant to hypoxia and hypoglycemia. Mechanistically, we demonstrated that BMP4 protected HCC cells from hypoxia and hypoglycemia by promoting glycolysis since BMP4 up-regulated glucose uptake, the lactic acid production, the ATP level, and the activities of rate limiting enzymes of glycolysis (including HK2, PFK and PK). Furthermore, we demonstrated that BMP4 up-regulated HK2, PFKFB3 and PKM2 through the canonical Smad signal pathway as SMAD5 directly bound to the promoter of PKM. Collectively, our findings shown that BMP4 may play an important role in regulating glycolysis of HCC cells under hypoxia and hypoglycemia condition, indicating that novel therapeutics may be developed to target BMP4-regulated glucose metabolic reprogramming in HCC.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Quan Kang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children’s Hospital of Chongqing Medical UniversityChongqing 400014, China
| | - Youde Cao
- Department of Pathology, Chongqing Medical UniversityChongqing 400016, China
| | - Baicheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical UniversityChongqing 400016, China
| | - Piao Zhao
- Ministry of Education Key Laboratory of Diagnostic Medicine, Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Yetao Luo
- Clinical Epidemiology and Biostatistics Department, Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical UniversityChongqing 400014, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical CenterChicago, IL 60637, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
8
|
Sha W, Hu F, Bu S. Mitochondrial dysfunction and pancreatic islet β-cell failure (Review). Exp Ther Med 2020; 20:266. [PMID: 33199991 PMCID: PMC7664595 DOI: 10.3892/etm.2020.9396] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cells are the only source of insulin in humans. Mitochondria uses pyruvate to produce ATP as an intermediate link between glucose intake and insulin secretion in β-cells, in a process known as glucose-stimulated insulin secretion (GSIS). Previous studies have demonstrated that GSIS is negatively regulated by various factors in the mitochondria, including tRNALeu mutations, high p58 expression, reduced nicotinamide nucleotide transhydrogenase activity, abnormal levels of uncoupling proteins and reduced expression levels of transcription factors A, B1 and B2. Additionally, oxidative stress damages mitochondria and impairs antioxidant defense mechanisms, leading to the increased production of reactive oxygen species, which induces β-cell dysfunction. Inflammation in islets can also damage β-cell physiology. Inflammatory cytokines trigger the release of cytochrome c from the mitochondria via the NF-κB pathway. The present review examined the potential factors underlying mitochondrial dysfunction and their association with islet β-cell failure, which may offer novel insights regarding future strategies for the preservation of mitochondrial function and enhancement of antioxidant activity for individuals with diabetes mellitus.
Collapse
Affiliation(s)
- Wenxin Sha
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Fei Hu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shizhong Bu
- Diabetes Research Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
9
|
Yin N, Xie T, Zhang H, Chen J, Yu J, Liu F. IDH1-R132H mutation radiosensitizes U87MG glioma cells via epigenetic downregulation of TIGAR. Oncol Lett 2019; 19:1322-1330. [PMID: 31966064 PMCID: PMC6956398 DOI: 10.3892/ol.2019.11148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/16/2019] [Indexed: 12/01/2022] Open
Abstract
Isocitrate dehydrogenase 1 (IDH1) is the most frequently mutated gene in World Health Organization grade II–III and secondary glioma. The majority of IDH1 mutation cases involve the substitution from arginine to histidine at codon 132 (IDH1-R132H). Although the oncogenic role of IDH1-R132H has been confirmed, patients with IDH1-R132H brain tumors exhibit a better response to radiotherapy compared with those with wild-type (WT) IDH1. In the present study, the potential mechanism of radiosensitization mediated by IDH1-R132H was investigated by overexpressing IDH1-R132H in U87MG glioma cells. The results demonstrated decreased clonogenic capacity of IDH1-R132H-expressing cells, as well as delayed repair of DNA double-strand breaks compared with IDH1-WT. Data from The Cancer Genome Atlas were analyzed, which demonstrated that the expression of TP53-induced glycolysis and apoptosis regulator (TIGAR) was lower in patients with glioma harboring IDH1 mutations compared with that in patients with IDH1-WT. TIGAR-knockdown increases the radiosensitivity of glioma cells; in U87MG cells, IDH1-R132H suppressed TIGAR expression. Chromatin immunoprecipitation assays revealed increased levels of repressive H3K9me3 markers at the TIGAR promoter in IDH1-R132H compared with IDH1-WT. These data indicated that IDH1-R132H may overcome radioresistance in glioma cells through epigenetic suppression of TIGAR expression. However, these favorable effects were not observed in U87MG glioma stem-like cells. The results of the present study provide an improved understanding of the functionality of IDH1 mutations in glioma cells, which may improve the therapeutic efficacy of radiotherapy.
Collapse
Affiliation(s)
- Narui Yin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| | - Ting Xie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| | - Haowen Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| | - Jian Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| | - Jiahua Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| | - Fenju Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China.,Department of Radiobiology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
10
|
Gelman SJ, Naser F, Mahieu NG, McKenzie LD, Dunn GP, Chheda MG, Patti GJ. Consumption of NADPH for 2-HG Synthesis Increases Pentose Phosphate Pathway Flux and Sensitizes Cells to Oxidative Stress. Cell Rep 2019; 22:512-522. [PMID: 29320744 PMCID: PMC6053654 DOI: 10.1016/j.celrep.2017.12.050] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 01/21/2023] Open
Abstract
Gain-of-function mutations in isocitrate dehydroge-nase 1 (IDH1) occur in multiple types of human cancer. Here, we show that these mutations significantly disrupt NADPH homeostasis by consuming NADPH for 2-hydroxyglutarate (2-HG) synthesis. Cells respond to 2-HG synthesis, but not exogenous administration of 2-HG, by increasing pentose phosphate pathway (PPP) flux. We show that 2-HG production competes with reductive biosynthesis and the buffering of oxidative stress, processes that also require NADPH. IDH1 mutants have a decreased capacity to synthesize palmitate and an increased sensitivity to oxidative stress. Our results demonstrate that, even when NADPH is limiting, IDH1 mutants continue to synthesize 2-HG at the expense of other NADPH-requiring pathways that are essential for cell viability. Thus, rather than attempting to decrease 2-HG synthesis in the clinic, the consumption of NADPH by mutant IDH1 may be exploited as a metabolic weakness that sensitizes tumor cells to ionizing radiation, a commonly used anti-cancer therapy.
Collapse
Affiliation(s)
- Susan J Gelman
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Fuad Naser
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Nathaniel G Mahieu
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Lisa D McKenzie
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gavin P Dunn
- Departments of Neurological Surgery and Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Milan G Chheda
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
Radiosensitization and a Less Aggressive Phenotype of Human Malignant Glioma Cells Expressing Isocitrate Dehydrogenase 1 (IDH1) Mutant Protein: Dissecting the Mechanisms. Cancers (Basel) 2019; 11:cancers11060889. [PMID: 31242696 PMCID: PMC6627228 DOI: 10.3390/cancers11060889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023] Open
Abstract
The presence of an isocitrate dehydrogenase 1 (IDH1) mutation is associated with a less aggressive phenotype, increased sensitivity to radiation, and increased overall survival in patients with diffuse glioma. Based on in vitro experimentations in malignant glioma cell lines, the consequences on cellular processes of IDH1R132H expression were analyzed. The results revealed that IDH1R132H expression enhanced the radiation induced accumulation of residual γH2AX foci and decreased the amount of glutathione (GSH) independent of the oxygen status. In addition, expression of the mutant IDH1 caused a significant increase of cell stiffness and induced an altered organization of the cytoskeleton, which has been shown to reinforce cell stiffness. Furthermore, IDH1R132H expression decreased the expression of vimentin, an important component of the cytoskeleton and regulator of the cell stiffness. The results emphasize the important role of mutant IDH1 in treatment of patients with diffuse gliomas especially in response to radiation. Hence, detection of the genetic status of IDH1 before therapy massively expands the utility of immunohistochemistry to accurately distinguish patients with a less aggressive and radiosensitive IDH1-mutant diffuse glioma suitable for radiotherapy from those with a more aggressive IDH1-wildtype diffuse glioma who might benefit from an individually intensified therapy comprising radiotherapy and alternative medical treatments.
Collapse
|
12
|
Molenaar RJ, Maciejewski JP, Wilmink JW, van Noorden CJF. Wild-type and mutated IDH1/2 enzymes and therapy responses. Oncogene 2018; 37:1949-1960. [PMID: 29367755 PMCID: PMC5895605 DOI: 10.1038/s41388-017-0077-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/14/2022]
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH1/2) are key enzymes in cellular metabolism, epigenetic regulation, redox states, and DNA repair. IDH1/2 mutations are causal in the development and/or progression of various types of cancer due to supraphysiological production of d-2-hydroxyglutarate. In various tumor types, IDH1/2-mutated cancers predict for improved responses to treatment with irradiation or chemotherapy. The present review discusses the molecular basis of the sensitivity of IDH1/2-mutated cancers with respect to the function of mutated IDH1/2 in cellular processes and their interactions with novel IDH1/2-mutant inhibitors. Finally, lessons learned from IDH1/2 mutations for future clinical applications in IDH1/2 wild-type cancers are discussed.
Collapse
Affiliation(s)
- Remco J Molenaar
- Cancer Center Amsterdam, Department of Medical Biology, Academic Medical Center, Amsterdam, The Netherlands. .,Cancer Center Amsterdam, Department of Medical Oncology, Academic Medical Center, Amsterdam, The Netherlands. .,Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA.
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Department of Medical Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Cancer Center Amsterdam, Department of Medical Biology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Kim S, Kim JE, Kim YH, Hwang T, Kim SK, Xu WJ, Shin JY, Kim JI, Choi H, Kim HC, Cho HR, Choi A, Chowdhury T, Seo Y, Dho YS, Kim JW, Kim DG, Park SH, Kim H, Choi SH, Park S, Lee SH, Park CK. Glutaminase 2 expression is associated with regional heterogeneity of 5-aminolevulinic acid fluorescence in glioblastoma. Sci Rep 2017; 7:12221. [PMID: 28939850 PMCID: PMC5610329 DOI: 10.1038/s41598-017-12557-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/12/2017] [Indexed: 01/11/2023] Open
Abstract
Fluorescence-guided surgery using 5-aminolevulinic acid (5-ALA) is now a widely-used modality for glioblastoma (GBM) treatment. However, intratumoral heterogeneity of fluorescence intensity may reflect different onco-metabolic programs. Here, we investigated the metabolic mechanism underlying the heterogeneity of 5-ALA fluorescence in GBM. Using an in-house developed fluorescence quantification system for tumor tissues, we collected 3 types of GBM tissues on the basis of their fluorescence intensity, which was characterized as strong, weak, and none. Expression profiling by RNA-sequencing revealed 77 genes with a proportional relationship and 509 genes with an inverse relationship between gene expression and fluorescence intensity. Functional analysis and in vitro experiments confirmed glutaminase 2 (GLS2) as a key gene associated with the fluorescence heterogeneity. Subsequent metabolite profiling discovered that insufficient NADPH due to GLS2 underexpression was responsible for the delayed metabolism of 5-ALA and accumulation of protoporphyrin IX (PpIX) in the high fluorescence area. The expression level of GLS2 and related NADPH production capacity is associated with the regional heterogeneity of 5-ALA fluorescence in GBM.
Collapse
Affiliation(s)
- Sojin Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Ja Eun Kim
- Cell and Gene Therapy Products Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Yong Hwy Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Taeyoung Hwang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sung Kwon Kim
- Department of Neurosurgery, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Wen Jun Xu
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, Korea
| | - Jong-Yeon Shin
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Jong-Il Kim
- Department of Biochemical and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | | | - Hee Chan Kim
- Department of Biomedical Engineering, College of Medicine and Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul, Korea
| | - Hye Rim Cho
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Anna Choi
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tamrin Chowdhury
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Youngbeom Seo
- Department of Neurosurgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Yun-Sik Dho
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyeonjin Kim
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Seoul, Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
14
|
Kim JE, Cho HR, Xu WJ, Kim JY, Kim SK, Kim SK, Park SH, Kim H, Lee SH, Choi SH, Park S, Park CK. Mechanism for enhanced 5-aminolevulinic acid fluorescence in isocitrate dehydrogenase 1 mutant malignant gliomas. Oncotarget 2016; 6:20266-77. [PMID: 26008980 PMCID: PMC4653003 DOI: 10.18632/oncotarget.4060] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/22/2015] [Indexed: 11/25/2022] Open
Abstract
Fluorescence-guided surgery using 5-aminolevulinic acid (5-ALA) has become the main treatment modality in malignant gliomas. However unlike glioblastomas, there are inconsistent result about fluorescence status in WHO grade III gliomas. Here, we show that mutational status of IDH1 is linked to 5-ALA fluorescence. Using genetically engineered malignant glioma cells harboring wild type (U87MG-IDH1WT) or mutant (U87MG-IDH1R132H) IDH1, we demonstrated a lag in 5-ALA metabolism and accumulation of protoporphyrin IX (PpIX) in U87MG-IDH1R132H cells. Next, we used liquid chromatography–mass spectrometry (LC-MS) to screen for tricarboxylic acid (TCA) cycle-related metabolite changes caused by 5-ALA exposure. We observed low baseline levels of NADPH, an essential cofactor for the rate-limiting step of heme degradation, in U87MG-IDH1R132H cells. High levels of NADPH are required to metabolize excessive 5-ALA, giving a plausible reason for the temporarily enhanced 5-ALA fluorescence in mutant IDH1 cells. This hypothesis was supported by the results of metabolic screening in human malignant glioma samples. In conclusion, we have discovered a relationship between enhanced 5-ALA fluorescence and IDH1 mutations in WHO grade III gliomas. Low levels of NADPH in tumors with mutated IDH1 is responsible for the enhanced fluorescence.
Collapse
Affiliation(s)
- Ja Eun Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Hye Rim Cho
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Wen Jun Xu
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Sillim-dong, Gwanak-gu, Seoul, Korea
| | - Ji Young Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Sung Kwon Kim
- Department of Neurosurgery, Gyeongsang National University School of Medicine, Gyeongsang National University Hospital, Jinju, Korea
| | - Seung-Ki Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea.,Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Jongno-gu, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Hyeonjin Kim
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Product Research Institute, Seoul National University, Sillim-dong, Gwanak-gu, Seoul, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| |
Collapse
|
15
|
Ilkhanizadeh S, Lau J, Huang M, Foster DJ, Wong R, Frantz A, Wang S, Weiss WA, Persson AI. Glial progenitors as targets for transformation in glioma. Adv Cancer Res 2015; 121:1-65. [PMID: 24889528 DOI: 10.1016/b978-0-12-800249-0.00001-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioma is the most common primary malignant brain tumor and arises throughout the central nervous system. Recent focus on stem-like glioma cells has implicated neural stem cells (NSCs), a minor precursor population restricted to germinal zones, as a potential source of gliomas. In this review, we focus on the relationship between oligodendrocyte progenitor cells (OPCs), the largest population of cycling glial progenitors in the postnatal brain, and gliomagenesis. OPCs can give rise to gliomas, with signaling pathways associated with NSCs also playing key roles during OPC lineage development. Gliomas can also undergo a switch from progenitor- to stem-like phenotype after therapy, consistent with an OPC-origin even for stem-like gliomas. Future in-depth studies of OPC biology may shed light on the etiology of OPC-derived gliomas and reveal new therapeutic avenues.
Collapse
Affiliation(s)
- Shirin Ilkhanizadeh
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Jasmine Lau
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Miller Huang
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Daniel J Foster
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - Robyn Wong
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Aaron Frantz
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - Susan Wang
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, California, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Department of Neurology, University of California, San Francisco, California, USA
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, California, USA; Department of Neurological Surgery and Brain Tumor Research Center, University of California, San Francisco, California, USA; Sandler Neurosciences Center, University of California, San Francisco, California, USA.
| |
Collapse
|
16
|
Chakrabarti G, Gerber DE, Boothman DA. Expanding antitumor therapeutic windows by targeting cancer-specific nicotinamide adenine dinucleotide phosphate-biogenesis pathways. Clin Pharmacol 2015; 7:57-68. [PMID: 25870517 PMCID: PMC4381889 DOI: 10.2147/cpaa.s79760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) biogenesis is an essential mechanism by which both normal and cancer cells maintain redox balance. While antitumor approaches to treat cancers through elevated reactive oxygen species (ROS) are not new ideas, depleting specific NADPH-biogenesis pathways that control recovery and repair pathways are novel, viable approaches to enhance cancer therapy. However, to elicit efficacious therapies exploiting NADPH-biogenic pathways, it is crucial to understand and specifically define the roles of NADPH-biogenesis pathways used by cancer cells for survival or recovery from cell stress. It is equally important to select NADPH-biogenic pathways that are expendable or not utilized in normal tissue to avoid unwanted toxicity. Here, we address recent literature that demonstrates specific tumor-selective NADPH-biogenesis pathways that can be exploited using agents that target specific cancer cell pathways normally not utilized in normal cells. Defining NADPH-biogenesis profiles of specific cancer-types should enable novel strategies to exploit these therapeutic windows for increased efficacy against recalcitrant neoplastic disease, such as pancreatic cancers. Accomplishing the goal of using ROS as a weapon against cancer cells will also require agents, such as NQO1 bioactivatable drugs, that selectively induce elevated ROS levels in cancer cells, while normal cells are protected.
Collapse
Affiliation(s)
- Gaurab Chakrabarti
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA ; Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - David E Gerber
- Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - David A Boothman
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA ; Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA ; Harold C Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
17
|
Hochberg FH, Atai NA, Gonda D, Hughes MS, Mawejje B, Balaj L, Carter RS. Glioma diagnostics and biomarkers: an ongoing challenge in the field of medicine and science. Expert Rev Mol Diagn 2014; 14:439-52. [PMID: 24746164 DOI: 10.1586/14737159.2014.905202] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioma is the most common brain tumor. For the more aggressive form, glioblastoma, standard treatment includes surgical resection, irradiation with adjuvant temozolomide and, on recurrence, experimental chemotherapy. However, the survival of patients remains poor. There is a critical need for minimally invasive biomarkers for diagnosis and as measures of response to therapeutic interventions. Glioma shed extracellular vesicles (EVs), which invade the surrounding tissue and circulate within both the cerebrospinal fluid and the systemic circulation. These tumor-derived EVs and their content serve as an attractive source of biomarkers. In this review, we discuss the current state of the art of biomarkers for glioma with emphasis on their EV derivation.
Collapse
Affiliation(s)
- Fred H Hochberg
- Department of Neurology and Program in Neuroscience, Massachusetts General Hospital and Harvard Medical School, Suite 340, 175 Cambridge Street, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Expression analysis of all protease genes reveals cathepsin K to be overexpressed in glioblastoma. PLoS One 2014; 9:e111819. [PMID: 25356585 PMCID: PMC4214761 DOI: 10.1371/journal.pone.0111819] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/01/2014] [Indexed: 01/26/2023] Open
Abstract
Background Cancer genome and transcriptome analyses advanced our understanding of cancer biology. We performed transcriptome analysis of all known genes of peptidases also called proteases and their endogenous inhibitors in glioblastoma multiforme (GBM), which is one of the most aggressive and deadly types of brain cancers, where unbalanced proteolysis is associated with tumor progression. Methods Comparisons were performed between the transcriptomics of primary GBM tumors and unmatched non-malignant brain tissue, and between GBM cell lines (U87-MG and U373) and a control human astrocyte cell line (NHA). Publicly-available data sets and our own datasets were integrated and normalized using bioinformatics tools to reveal protease and protease inhibitor genes with deregulated expression in both malignant versus non-malignant tissues and cells. Results Of the 311 protease genes identified to be differentially expressed in both GBM tissues and cells, 5 genes were highly overexpressed, 2 genes coding for non-peptidase homologues transferrin receptor (TFRC) and G protein-coupled receptor 56 (GPR56), as well as 3 genes coding for the proteases endoplasmic reticulum aminopeptidase 2 (ERAP2), glutamine-fructose-6-phosphate transaminase 2 (GFPT2) and cathepsin K (CTSK), whereas one gene, that of the serine protease carboxypeptidase E (CPE) was strongly reduced in expression. Seventy five protease inhibitor genes were differentially expressed, of which 3 genes were highly overexpressed, the genes coding for stefin B (CSTB), peptidase inhibitor 3 (PI3 also named elafin) and CD74. Seven out of 8 genes (except CSTB) were validated using RT-qPCR in GBM cell lines. CTSK overexpression was validated using RT-qPCR in GBM tissues as well. Cathepsin K immunohistochemical staining and western blotting showed that only proteolytically inactive proforms of cathepsin K were overexpressed in GBM tissues and cells. Conclusions The presence of high levels of inactive proforms of cathepsin K in GBM tissues and cells indicate that in GBM the proteolytic/collagenolytic role is not its primary function but it plays rather a different yet unknown role.
Collapse
|
19
|
Lynes J, Wibowo M, Koschmann C, Baker GJ, Saxena V, Muhammad AKMG, Bondale N, Klein J, Assi H, Lieberman AP, Castro MG, Lowenstein PR. Lentiviral-induced high-grade gliomas in rats: the effects of PDGFB, HRAS-G12V, AKT, and IDH1-R132H. Neurotherapeutics 2014; 11:623-35. [PMID: 24752661 PMCID: PMC4121445 DOI: 10.1007/s13311-014-0269-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In human gliomas, the RTK/RAS/PI(3)K signaling pathway is nearly always altered. We present a model of experimental gliomagenesis that elucidates the contributions of genes involved in this pathway (PDGF-B ligand, HRAS-G12V, and AKT). We also examine the effect on gliomagenesis by the potential modifier gene, IDH1-R132H. Injections of lentiviral-encoded oncogenes induce de novo gliomas of varying penetrance, tumor progression, and histological grade depending on the specific oncogenes used. Our model mimics hallmark histological structures of high-grade glioma, such as pseudopalisades, glomeruloid microvascular proliferation, and diffuse tumor invasion. We use our model of gliomagenesis to test the efficacy of an experimental brain tumor gene therapy. Our model allowed us to test the contributions of oncogenes in the RTK/RAS/PI(3)K pathway, and their potential modification by over-expression of mutated IDH1, in glioma development and progression in rats. Our model constitutes a clinically relevant system to study gliomagenesis, the effects of modifier genes, and the efficacy of experimental therapeutics.
Collapse
Affiliation(s)
- John Lynes
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Mia Wibowo
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Carl Koschmann
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Gregory J. Baker
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Vandana Saxena
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - A. K. M. G. Muhammad
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Niyati Bondale
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Julia Klein
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Hikmat Assi
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Andrew P. Lieberman
- />Department of Pathology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Maria G. Castro
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| | - Pedro R. Lowenstein
- />Department of Neurosurgery, University of Michigan, School of Medicine, 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109 USA
- />Department of Cell and Developmental Biology, University of Michigan, School of Medicine, Ann Arbor, MI 48109 USA
| |
Collapse
|
20
|
Molenaar RJ, Radivoyevitch T, Maciejewski JP, van Noorden CJF, Bleeker FE. The driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in oncogenesis and survival prolongation. Biochim Biophys Acta Rev Cancer 2014; 1846:326-41. [PMID: 24880135 DOI: 10.1016/j.bbcan.2014.05.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/30/2014] [Accepted: 05/22/2014] [Indexed: 01/06/2023]
Abstract
Mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are key events in the development of glioma, acute myeloid leukemia (AML), chondrosarcoma, intrahepatic cholangiocarcinoma (ICC), and angioimmunoblastic T-cell lymphoma. They also cause D-2-hydroxyglutaric aciduria and Ollier and Maffucci syndromes. IDH1/2 mutations are associated with prolonged survival in glioma and in ICC, but not in AML. The reason for this is unknown. In their wild-type forms, IDH1 and IDH2 convert isocitrate and NADP(+) to α-ketoglutarate (αKG) and NADPH. Missense mutations in the active sites of these enzymes induce a neo-enzymatic reaction wherein NADPH reduces αKG to D-2-hydroxyglutarate (D-2HG). The resulting D-2HG accumulation leads to hypoxia-inducible factor 1α degradation, and changes in epigenetics and extracellular matrix homeostasis. Such mutations also imply less NADPH production capacity. Each of these effects could play a role in cancer formation. Here, we provide an overview of the literature and discuss which downstream molecular effects are likely to be the drivers of the oncogenic and survival-prolonging properties of IDH1/2 mutations. We discuss interactions between mutant IDH1/2 inhibitors and conventional therapies. Understanding of the biochemical consequences of IDH1/2 mutations in oncogenesis and survival prolongation will yield valuable information for rational therapy design: it will tell us which oncogenic processes should be blocked and which "survivalogenic" effects should be retained.
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Cell Biology & Histology, Academic Medical Center, University of Amsterdam, The Netherlands.
| | - Tomas Radivoyevitch
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Cornelis J F van Noorden
- Department of Cell Biology & Histology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Fonnet E Bleeker
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
21
|
Van Noorden CJF. The NADP+-dependent IDH1 mutation and its relevance for glioblastoma patient survival. Cancer Metab 2014. [PMCID: PMC4073009 DOI: 10.1186/2049-3002-2-s1-p79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
22
|
Glutamate as chemotactic fuel for diffuse glioma cells: are they glutamate suckers? Biochim Biophys Acta Rev Cancer 2014; 1846:66-74. [PMID: 24747768 DOI: 10.1016/j.bbcan.2014.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 11/21/2022]
Abstract
Diffuse gliomas comprise a group of primary brain tumors that originate from glial (precursor) cells and present as a variety of malignancy grades which have in common that they grow by diffuse infiltration. This phenotype complicates treatment enormously as it precludes curative surgery and radiotherapy. Furthermore, diffusely infiltrating glioma cells often hide behind a functional blood-brain barrier, hampering delivery of systemically administered therapeutic and diagnostic compounds to the tumor cells. The present review addresses the biological mechanisms that underlie the diffuse infiltrative phenotype, knowledge of which may improve treatment strategies for this disastrous tumor type. The invasive phenotype is specific for glioma: most other brain tumor types, both primary and metastatic, grow as delineated lesions. Differences between the genetic make-up of glioma and that of other tumor types may therefore help to unravel molecular pathways, involved in diffuse infiltrative growth. One such difference concerns mutations in the NADP(+)-dependent isocitrate dehydrogenase (IDH1 and IDH2) genes, which occur in >80% of cases of low grade glioma and secondary glioblastoma. In this review we present a novel hypothesis which links IDH1 and IDH2 mutations to glutamate metabolism, possibly explaining the specific biological behavior of diffuse glioma.
Collapse
|
23
|
Van Noorden C. Imaging enzymes at work: metabolic mapping by enzyme histochemistry to study functional mechanisms of the isocitrate dehydrogenase IDH1 mutation in glioblastoma (59.3). FASEB J 2014. [DOI: 10.1096/fasebj.28.1_supplement.59.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
24
|
Navis AC, Niclou SP, Fack F, Stieber D, van Lith S, Verrijp K, Wright A, Stauber J, Tops B, Otte-Holler I, Wevers RA, van Rooij A, Pusch S, von Deimling A, Tigchelaar W, van Noorden CJF, Wesseling P, Leenders WPJ. Increased mitochondrial activity in a novel IDH1-R132H mutant human oligodendroglioma xenograft model: in situ detection of 2-HG and α-KG. Acta Neuropathol Commun 2013; 1:18. [PMID: 24252742 PMCID: PMC3893588 DOI: 10.1186/2051-5960-1-18] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/09/2013] [Indexed: 01/04/2023] Open
Abstract
Background Point mutations in genes encoding NADP+-dependent isocitrate dehydrogenases (especially IDH1) are common in lower grade diffuse gliomas and secondary glioblastomas and occur early during tumor development. The contribution of these mutations to gliomagenesis is not completely understood and research is hampered by the lack of relevant tumor models. We previously described the development of the patient-derived high-grade oligodendroglioma xenograft model E478 that carries the commonly occurring IDH1-R132H mutation. We here report on the analyses of E478 xenografts at the genetic, histologic and metabolic level. Results LC-MS and in situ mass spectrometric imaging by LESA-nano ESI-FTICR revealed high levels of the proposed oncometabolite D-2-hydroxyglutarate (D-2HG), the product of enzymatic conversion of α-ketoglutarate (α-KG) by IDH1-R132H, in the tumor but not in surrounding brain parenchyma. α-KG levels and total NADP+-dependent IDH activity were similar in IDH1-mutant and -wildtype xenografts, demonstrating that IDH1-mutated cancer cells maintain α-KG levels. Interestingly, IDH1-mutant tumor cells in vivo present with high densities of mitochondria and increased levels of mitochondrial activity as compared to IDH1-wildtype xenografts. It is not yet clear whether this altered mitochondrial activity is a driver or a consequence of tumorigenesis. Conclusions The oligodendroglioma model presented here is a valuable model for further functional elucidation of the effects of IDH1 mutations on tumor metabolism and may aid in the rational development of novel therapeutic strategies for the large subgroup of gliomas carrying IDH1 mutations.
Collapse
|
25
|
Horbinski C. What do we know about IDH1/2 mutations so far, and how do we use it? Acta Neuropathol 2013; 125:621-36. [PMID: 23512379 DOI: 10.1007/s00401-013-1106-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 03/09/2013] [Indexed: 12/16/2022]
Abstract
Whole genome analyses have facilitated the discovery of clinically relevant genetic alterations in a variety of diseases, most notably cancer. A prominent example of this was the discovery of mutations in isocitrate dehydrogenases 1 and 2 (IDH1/2) in a sizeable proportion of gliomas and some other neoplasms. Herein the normal functions of these enzymes, how the mutations alter their catalytic properties, the effects of their D-2-hydroxyglutarate metabolite, technical considerations in diagnostic neuropathology, implications about prognosis and therapeutic considerations, and practical applications and controversies regarding IDH1/2 mutation testing are discussed.
Collapse
Affiliation(s)
- Craig Horbinski
- Department of Pathology, University of Kentucky, 307 Combs Cancer Research Facility, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
26
|
Baldewpersad Tewarie NMS, Burgers IAV, Dawood Y, den Boon HC, den Brok MGHE, Klunder JH, Koopmans KB, Rademaker E, van den Broek HB, van den Bersselaar SM, Witjes JJ, Van Noorden CJF, Atai NA. NADP+ -dependent IDH1 R132 mutation and its relevance for glioma patient survival. Med Hypotheses 2013; 80:728-31. [PMID: 23541771 DOI: 10.1016/j.mehy.2013.02.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 02/23/2013] [Indexed: 01/06/2023]
Abstract
The isocitrate dehydrogenase 1 (IDH1) mutation occurs in high frequency in glioma and secondary glioblastoma (GBM). Mutated IDH1 produces the oncometabolite 2-hydroxyglutarate rather than α-ketoglutarate or isocitrate. The oncometabolite is considered to be the major cause of the association between the IDH1 mutation and gliomagenesis. On the other hand, the IDH1 mutation in GBM is associated with prolonged patient survival. This association is not well understood yet but IDH1 involvement in epigenetic silencing of O-6-methylguanine-DNA methyltransferase (MGMT), a DNA repair enzyme is considered to be an important mechanism. However, it was shown recently that the IDH1 mutation and MGMT silencing are independent prognostic factors. Here, we hypothesize that the IDH1 mutation reduces the capacity to produce NADPH and thus reduces the capacity to scavenge reactive oxygen species that are generated during irradiation and chemotherapy. IDH1 activity is responsible for two-thirds of the NADPH production capacity in normal brain, whereas the IDH1 mutation reduces this capacity by almost 40%. Therefore, we hypothesize that the reduced NADPH production capacity due to the IDH1 mutation renders GBM cells more vulnerable to irradiation and chemotherapy thus prolonging survival of the patients.
Collapse
|
27
|
Ward PS, Lu C, Cross JR, Abdel-Wahab O, Levine RL, Schwartz GK, Thompson CB. The potential for isocitrate dehydrogenase mutations to produce 2-hydroxyglutarate depends on allele specificity and subcellular compartmentalization. J Biol Chem 2012; 288:3804-15. [PMID: 23264629 DOI: 10.1074/jbc.m112.435495] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Monoallelic point mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) and its mitochondrial homolog IDH2 can lead to elevated levels of 2-hydroxyglutarate (2HG) in multiple cancers. Here we report that cellular 2HG production from cytosolic IDH1 mutation is dependent on the activity of a retained wild-type IDH1 allele. In contrast, expression of mitochondrial IDH2 mutations led to robust 2HG production in a manner independent of wild-type mitochondrial IDH function. Among the recurrent IDH2 mutations at Arg-172 and Arg-140, IDH2 Arg-172 mutations consistently led to greater 2HG accumulation than IDH2 Arg-140 mutations, and the degree of 2HG accumulation correlated with the ability of these mutations to block cellular differentiation. Cytosolic IDH1 Arg-132 mutations, although structurally analogous to mutations at mitochondrial IDH2 Arg-172, were only able to elevate intracellular 2HG to comparable levels when an equivalent level of wild-type IDH1 was co-expressed. Consistent with 2HG production from cytosolic IDH1 being limited by substrate production from wild-type IDH1, we observed 2HG levels to increase in cancer cells harboring an endogenous monoallelic IDH1 mutation when mitochondrial IDH flux was diverted to the cytosol. Finally, expression of an IDH1 construct engineered to localize to the mitochondria rather than the cytosol resulted in greater 2HG accumulation. These data demonstrate that allelic and subcellular compartment differences can regulate the potential for IDH mutations to produce 2HG in cells. The consequences of 2HG elevation are dose-dependent, and the non-equivalent 2HG accumulation resulting from IDH1 and IDH2 mutations may underlie their differential prognosis and prevalence in various cancers.
Collapse
Affiliation(s)
- Patrick S Ward
- Cancer Biology and Genetics Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Yang H, Ye D, Guan KL, Xiong Y. IDH1 and IDH2 mutations in tumorigenesis: mechanistic insights and clinical perspectives. Clin Cancer Res 2012; 18:5562-71. [PMID: 23071358 PMCID: PMC3897211 DOI: 10.1158/1078-0432.ccr-12-1773] [Citation(s) in RCA: 320] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genes encoding for isocitrate dehydrogenases 1 and 2, IDH1 and IDH2, are frequently mutated in multiple types of human cancer. Mutations targeting IDH1 and IDH2 result in simultaneous loss of their normal catalytic activity, the production of α-ketoglutarate (α-KG), and gain of a new function, the production of 2-hydroxyglutarate (2-HG). 2-HG is structurally similar to α-KG, and acts as an α-KG antagonist to competitively inhibit multiple α-KG-dependent dioxygenases, including both lysine histone demethylases and the ten-eleven translocation family of DNA hydroxylases. Abnormal histone and DNA methylation are emerging as a common feature of tumors with IDH1 and IDH2 mutations and may cause altered stem cell differentiation and eventual tumorigenesis. Therapeutically, unique features of IDH1 and IDH2 mutations make them good biomarkers and potential drug targets.
Collapse
Affiliation(s)
- Hui Yang
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences and School of Life Sciences, Fudan University, Shanghai, P R China
| | | | | | | |
Collapse
|
29
|
Rankin SL, Zhu G, Baker SJ. Review: insights gained from modelling high-grade glioma in the mouse. Neuropathol Appl Neurobiol 2012; 38:254-70. [PMID: 22035336 DOI: 10.1111/j.1365-2990.2011.01231.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High-grade gliomas (HGGs) are devastating primary brain tumours with poor outcomes. Advances towards effective treatments require improved understanding of pathogenesis and relevant model systems for preclinical testing. Mouse models for HGG provide physiologically relevant experimental systems for analysis of HGG pathogenesis. There are advantages and disadvantages to the different methodologies used to generate such models, including implantation, genetic engineering or somatic gene transfer approaches. This review highlights how mouse models have provided insights into the contribution of specific mutations to tumour initiation, progression and phenotype, the influence of tumour micro-environment, and the analysis of cell types that can give rise to glioma. HGGs are a heterogeneous group of tumours, and the complexity of diverse mutations within common signalling pathways as well as the developmental and cell-type context of transformation contributes to the overall diversity of glioma phenotype. Enhanced understanding of the mutations and cell types giving rise to HGG, along with the ability to design increasingly complex mouse models that more closely simulate the process of human gliomagenesis will continue to provide improved experimental systems for dissecting mechanisms of disease pathogenesis and for preclinical testing.
Collapse
Affiliation(s)
- S L Rankin
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | |
Collapse
|