1
|
Li W, Chen H, Yuan X, Yao Q, Zhang M. Study of the role of leukocyte telomere length-related lncRNA NBR2 in Alzheimer's disease. Aging (Albany NY) 2024; 16:12593-12607. [PMID: 39287993 PMCID: PMC11466486 DOI: 10.18632/aging.206107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 09/19/2024]
Abstract
Alzheimer's Syndrome (AD) is a neurodegenerative disease that is prevalent in middle-aged and elderly people. As the disease progresses, patients gradually lose the ability to take care of themselves, which brings a heavy burden to the family. There is a link between leukocyte telomere length (LTL) and cognitive ability. To search for possible pathogenic mechanisms and potential therapeutic agents, we demonstrated a causal link between LTL and AD using Mendelian randomization analysis (MR). The expression of the target gene NBR2 and the downstream mRNA GJA1 and GJA1-related genes, pathway enrichment, and association with immune cells were further explored. Using the gene cluster-drug target interaction network, we obtained potential therapeutic drugs. Our study provides evidence for a causal link between AD and LTL, suggesting medicines that may treat and alleviate AD symptoms.
Collapse
Affiliation(s)
- Wenjie Li
- Department of Geriatrics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Haoyan Chen
- Department of Geriatrics, Jiangsu Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiaofan Yuan
- Department of Radiology of the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Qi Yao
- Department of Geriatrics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Mingjiong Zhang
- Department of Geriatrics, Jiangsu Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
2
|
Papageorgakopoulou MA, Bania A, Lagogianni IA, Birmpas K, Assimakopoulou M. The Role of Glia Telomere Dysfunction in the Pathogenesis of Central Nervous System Diseases. Mol Neurobiol 2024; 61:5868-5881. [PMID: 38240992 PMCID: PMC11249767 DOI: 10.1007/s12035-024-03947-6] [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: 10/10/2023] [Accepted: 01/09/2024] [Indexed: 07/16/2024]
Abstract
Maintaining the telomere length is decisive for the viability and homeostasis process of all the cells of an organism, including human glial cells. Telomere shortening of microglial cells has been widely associated with the onset and progression of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Additionally, traumatic brain injury appears to have a positive correlation with the telomere-shortening process of microglia, and telomere length can be used as a non-invasive biomarker for the clinical management of these patients. Moreover, telomere involvement through telomerase reactivation and homologous recombination also known as the alternative lengthening of telomeres (ALT) has been described in gliomagenesis pathways, and particular focus has been given in the translational significance of these mechanisms in gliomas diagnosis and prognostic classification. Finally, glia telomere shortening is implicated in some psychiatric diseases. Given that telomere dysfunction of glial cells is involved in the central nervous system (CNS) disease pathogenesis, it represents a promising drug target that could lead to the incorporation of new tools in the medicinal arsenal for the management of so far incurable conditions.
Collapse
Affiliation(s)
| | - Angelina Bania
- School of Medicine, University of Patras, 26504, Patras, Greece
| | | | | | - Martha Assimakopoulou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Preclinical Medicine Department Building, 1 Asklipiou, 26504, Patras, Greece.
| |
Collapse
|
3
|
Rudà R, Horbinski C, van den Bent M, Preusser M, Soffietti R. IDH inhibition in gliomas: from preclinical models to clinical trials. Nat Rev Neurol 2024; 20:395-407. [PMID: 38760442 DOI: 10.1038/s41582-024-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Gliomas are the most common malignant primary brain tumours in adults and cannot usually be cured with standard cancer treatments. Gliomas show intratumoural and intertumoural heterogeneity at the histological and molecular levels, and they frequently contain mutations in the isocitrate dehydrogenase 1 (IDH1) or IDH2 gene. IDH-mutant adult-type diffuse gliomas are subdivided into grade 2, 3 or 4 IDH-mutant astrocytomas and grade 2 or 3 IDH-mutant, 1p19q-codeleted oligodendrogliomas. The product of the mutated IDH genes, D-2-hydroxyglutarate (D-2-HG), induces global DNA hypermethylation and interferes with immunity, leading to stimulation of tumour growth. Selective inhibitors of mutant IDH, such as ivosidenib and vorasidenib, have been shown to reduce D-2-HG levels and induce cellular differentiation in preclinical models and to induce MRI-detectable responses in early clinical trials. The phase III INDIGO trial has demonstrated superiority of vorasidenib, a brain-penetrant pan-mutant IDH inhibitor, over placebo in people with non-enhancing grade 2 IDH-mutant gliomas following surgery. In this Review, we describe the pathway of development of IDH inhibitors in IDH-mutant low-grade gliomas from preclinical models to clinical trials. We discuss the practice-changing implications of the INDIGO trial and consider new avenues of investigation in the field of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy.
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Martin van den Bent
- Brain Tumour Center at Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy
| |
Collapse
|
4
|
Drobyshev A, Modestov A, Suntsova M, Poddubskaya E, Seryakov A, Moisseev A, Sorokin M, Tkachev V, Zakharova G, Simonov A, Zolotovskaia MA, Buzdin A. Pan-cancer experimental characteristic of human transcriptional patterns connected with telomerase reverse transcriptase ( TERT) gene expression status. Front Genet 2024; 15:1401100. [PMID: 38859942 PMCID: PMC11163056 DOI: 10.3389/fgene.2024.1401100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
The TERT gene encodes the reverse transcriptase subunit of telomerase and is normally transcriptionally suppressed in differentiated human cells but reactivated in cancers where its expression is frequently associated with poor survival prognosis. Here we experimentally assessed the RNA sequencing expression patterns associated with TERT transcription in 1039 human cancer samples of 27 tumor types. We observed a bimodal distribution of TERT expression where ∼27% of cancer samples did not express TERT and the rest showed a bell-shaped distribution. Expression of TERT strongly correlated with 1443 human genes including 103 encoding transcriptional factor proteins. Comparison of TERT- positive and negative cancers showed the differential activation of 496 genes and 1975 molecular pathways. Therein, 32/38 (84%) of DNA repair pathways were hyperactivated in TERT+ cancers which was also connected with accelerated replication, transcription, translation, and cell cycle progression. In contrast, the level of 40 positive cell cycle regulator proteins and a set of epithelial-to-mesenchymal transition pathways was specific for the TERT- group suggesting different proliferation strategies for both groups of cancer. Our pilot study showed that the TERT+ group had ∼13% of cancers with C228T or C250T mutated TERT promoter. However, the presence of promoter mutations was not associated with greater TERT expression compared with other TERT+ cancers, suggesting parallel mechanisms of its transcriptional activation in cancers. In addition, we detected a decreased expression of L1 retrotransposons in the TERT+ group, and further decreased L1 expression in promoter mutated TERT+ cancers. TERT expression was correlated with 17 genes encoding molecular targets of cancer therapeutics and may relate to differential survival patterns of TERT- positive and negative cancers.
Collapse
Affiliation(s)
- Aleksey Drobyshev
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Modestov
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Suntsova
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elena Poddubskaya
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Clinical Center Vitamed, Moscow, Russia
| | | | - Aleksey Moisseev
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maksim Sorokin
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Galina Zakharova
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksander Simonov
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marianna A. Zolotovskaia
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Center for Advanced Studies 20, Moscow, Russia
| | - Anton Buzdin
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Center for Advanced Studies 20, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
5
|
Bedics G, Szőke P, Bátai B, Nagy T, Papp G, Kránitz N, Rajnai H, Reiniger L, Bödör C, Scheich B. Novel, clinically relevant genomic patterns identified by comprehensive genomic profiling in ATRX-deficient IDH-wildtype adult high-grade gliomas. Sci Rep 2023; 13:18436. [PMID: 37891325 PMCID: PMC10611758 DOI: 10.1038/s41598-023-45786-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023] Open
Abstract
Glioblastomas are the most common IDH-wildtype adult high-grade gliomas, frequently harboring mutations in the TERT gene promoter (pTERT) and utilizing the subsequent telomerase overexpression for telomere length maintenance. However, some rare cases show loss of ATRX and use alternative mechanisms of telomere lengthening. In this study, we performed the first complex genomic analysis specifically concentrating on the latter subgroup. Comprehensive genomic profiling of 12 ATRX-deficient and 13 ATRX-intact IDH-wildtype adult high-grade gliomas revealed that ATRX and pTERT mutations are mutually exclusive. DNMT3A alterations were confined to ATRX-deficient, while PTEN mutations to ATRX-intact cases. RAS-MAPK pathway alterations, including NF1 mutations, were more characteristic in the ATRX-deficient group. Variants of genes related to homologous recombination repair showed different patterns of affected genes. Two ATRX-deficient tumors with high tumor mutational burden and mismatch repair deficiency were found. One of these contained a novel fusion involving the NTRK2 and LRRFIP2 genes, while the other showed loss of MSH2 and MSH6 without genetic alterations in the encoding genes suggesting an epigenetic background. Genetic characteristics of ATRX-deficient IDH-wildtype adult high-grade gliomas suggest that these tumors are particularly intriguing targets of potential future therapeutic interventions including immunotherapies combined with MAPK pathway inhibition and DNA repair inhibitors.
Collapse
Affiliation(s)
- Gábor Bedics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Péter Szőke
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Bence Bátai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Tibor Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Life Science Building, Debrecen, 4032, Hungary
| | - Gergő Papp
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Noémi Kránitz
- Department of Pathology, County Hospital Győr, Petz Aladár Hospital, Vasvári Pál út 2-4, Győr, 9024, Hungary
| | - Hajnalka Rajnai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Lilla Reiniger
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- HCEMM-SE Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary.
| |
Collapse
|
6
|
Chojak R, Fares J, Petrosyan E, Lesniak MS. Cellular senescence in glioma. J Neurooncol 2023; 164:11-29. [PMID: 37458855 DOI: 10.1007/s11060-023-04387-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/01/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Glioma is the most common primary brain tumor and is often associated with treatment resistance and poor prognosis. Standard treatment typically involves radiotherapy and temozolomide-based chemotherapy, both of which induce cellular senescence-a tumor suppression mechanism. DISCUSSION Gliomas employ various mechanisms to bypass or escape senescence and remain in a proliferative state. Importantly, senescent cells remain viable and secrete a large number of factors collectively known as the senescence-associated secretory phenotype (SASP) that, paradoxically, also have pro-tumorigenic effects. Furthermore, senescent cells may represent one form of tumor dormancy and play a role in glioma recurrence and progression. CONCLUSION In this article, we delineate an overview of senescence in the context of gliomas, including the mechanisms that lead to senescence induction, bypass, and escape. Furthermore, we examine the role of senescent cells in the tumor microenvironment and their role in tumor progression and recurrence. Additionally, we highlight potential therapeutic opportunities for targeting senescence in glioma.
Collapse
Affiliation(s)
- Rafał Chojak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Edgar Petrosyan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA.
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
7
|
Lee K, Kim SI, Kim EE, Shim YM, Won JK, Park CK, Choi SH, Yun H, Lee H, Park SH. Genomic profiles of IDH-mutant gliomas: MYCN-amplified IDH-mutant astrocytoma had the worst prognosis. Sci Rep 2023; 13:6761. [PMID: 37185778 PMCID: PMC10130138 DOI: 10.1038/s41598-023-32153-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to find any ambiguous genetic outlier for "oligodendroglioma, IDH-mutant and 1p/19q-codeleted (O_IDH_mut)" and "astrocytoma, IDH-mutant (A_IDH_mut)" and to redefine the genetic landscape and prognostic factors of IDH-mutant gliomas. Next-generation sequencing (NGS) using a brain tumor-targeted gene panel, methylation profiles, and clinicopathological features were analyzed for O_IDH_mut (n = 74) in 70 patients and for A_IDH_mut (n = 95) in 90 patients. 97.3% of O_IDH_mut and 98.9% of A_IDH_mut displayed a classic genomic landscape. Combined CIC (75.7%) and/or FUBP1 (45.9%) mutations were detected in 93.2% and MGMTp methylation in 95.9% of O_IDH_mut patients. In A_IDH_mut, TP53 mutations were found in 86.3% and combined ATRX (82.1%) and TERTp (6.3%) mutations in 88.4%. Although there were 3 confusing cases, NOS (not otherwise specified) category, based on genetic profiles, but they were clearly classified by combining histopathology and DKFZ methylation classifier algorithms. The patients with MYCN amplification and/or CDKN2A/2B homozygous deletion in the A_IDH_mut category had a worse prognosis than those without these gene alterations and MYCN-amplified A_IDH_mut showed the worst prognosis. However, there was no prognostic genetic marker in O_IDH_mut. In histopathologically or genetically ambiguous cases, methylation profiles can be used as an objective tool to avoid a diagnosis of NOS or NEC (not elsewhere classified), as well as for tumor classification. The authors have not encountered a case of true mixed oligoastrocytoma using an integrated diagnosis of histopathological, genetic and methylation profiles. MYCN amplification, in addition to CDKN2A/2B homozygous deletion, should be included in the genetic criteria for CNS WHO grade 4 A_IDH_mut.
Collapse
Affiliation(s)
- Kwanghoon Lee
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seong-Ik Kim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Eric Eunshik Kim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yu-Mi Shim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Artificial Intelligence Graduate School, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Bader JM, Deigendesch N, Misch M, Mann M, Koch A, Meissner F. Proteomics separates adult-type diffuse high-grade gliomas in metabolic subgroups independent of 1p/19q codeletion and across IDH mutational status. Cell Rep Med 2023; 4:100877. [PMID: 36584682 PMCID: PMC9873829 DOI: 10.1016/j.xcrm.2022.100877] [Citation(s) in RCA: 4] [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: 08/25/2021] [Revised: 07/15/2022] [Accepted: 12/07/2022] [Indexed: 12/30/2022]
Abstract
High-grade adult-type diffuse gliomas are malignant neuroepithelial tumors with poor survival rates in combined chemoradiotherapy. The current WHO classification is based on IDH1/2 mutational and 1p/19q codeletion status. Glioma proteome alterations remain undercharacterized despite their promise for a better molecular patient stratification and therapeutic target identification. Here, we use mass spectrometry to characterize 42 formalin-fixed, paraffin-embedded (FFPE) samples from IDH-wild-type (IDHwt) gliomas, IDH-mutant (IDHmut) gliomas with and without 1p/19q codeletion, and non-neoplastic controls. Based on more than 5,500 quantified proteins and 5,000 phosphosites, gliomas separate by IDH1/2 mutational status but not by 1p/19q status. Instead, IDHmut gliomas split into two proteomic subtypes with widespread perturbations, including aerobic/anaerobic energy metabolism. Validations with three independent glioma proteome datasets confirm these subgroups and link the IDHmut subtypes to the established proneural and classic/mesenchymal subtypes in IDHwt glioma. This demonstrates common phenotypic subtypes across the IDH status with potential therapeutic implications for patients with IDHmut gliomas.
Collapse
Affiliation(s)
- Jakob Maximilian Bader
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Martin Misch
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arend Koch
- Department of Neuropathology, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany.
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Department of Systems Immunology and Proteomics, Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
9
|
Shang J, Wang Y, Li Z, Jiang L, Bai Q, Zhang X, Xiao G, Zhang J. ATRX-dependent SVCT2 mediates macrophage infiltration in the glioblastoma xenograft model. J Neurophysiol 2022; 127:1309-1316. [PMID: 35417255 DOI: 10.1152/jn.00486.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alpha thalassemia/mental retardation syndrome X-linked (ATRX) mutation impairs DNA damage repair in glioblastoma (GBM), making these cells more susceptible to treatment, which may contribute to the survival advantage in GBM patients containing ATRX mutations. To better understand the role of ATRX in GBM, genes correlated with ATRX expression were screened in the Cancer Genome Atlas (702 cases) and Chinese Glioma Genome Atlas (325 cases) databases. Sodium-vitamin C cotransporter 2 (SVCT2) was the most positively correlated gene with ATRX expression. ATRX (about 1.99-fold) and SVCT2 (about 2.25-fold) were upregulated in GBM tissues from 40 patients compared to normal brain tissues from 23 subjects. ShSVCT2 transfection did not alter the in vitro viability of GL261 cells. At the same time, it could inhibit the proliferation of GL261 cells in the orthotopic transplantation model with diminished infiltrating macrophages (CD45highCD11b+), down-regulated chemokine (C-C motif) ligand 2 (Ccl2), Ccl4, C-X-C motif chemokine ligand 1 (Cxcl1), and Cxcl15 expression, and decreased p-IκBα and p-c-Jun expression. Effect of ShSVCT2 transfection could be reversed by overexpression of SVCT2. siRNA interference of ATRX-dependent SVCT2 signal with shSVCT2 could inhibit tumor cell proliferation in Glu261-LuNeo xenograft tumor model with more survival advantage, probably by the inhibited macrophage chemotaxis. These results indicate that ATRX-dependent SVCT2-mediated chemokine-induced macrophage infiltration is regulated by the NF-κB pathway, which could be considered as treatment targets.
Collapse
Affiliation(s)
- Jinxing Shang
- Department of Neurosurgery, grid.452270.6Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yana Wang
- Cangzhou Medical College, Cangzhou Higher Education District, Hebei Province, Cangzhou, Hebei, China
| | - Zhuangzhuang Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lijun Jiang
- Department of Neurosurgery, grid.452270.6Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Qingling Bai
- Department of Neurosurgery, grid.452270.6Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Xiaoling Zhang
- Department of Pathology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Guoxin Xiao
- Department of Neurosurgery, Cangxian Hospital, Cangzhou, Hebei, China
| | - Jinguo Zhang
- Department of Neurology, Mengcun County Hospital, Mengcun County, Cangzhou, Hebei, China
| |
Collapse
|
10
|
Taglang C, Batsios G, Mukherjee J, Tran M, Gillespie AM, Hong D, Ronen SM, Artee Luchman H, Pieper RO, Viswanath P. Deuterium magnetic resonance spectroscopy enables noninvasive metabolic imaging of tumor burden and response to therapy in low-grade gliomas. Neuro Oncol 2022; 24:1101-1112. [PMID: 35091751 PMCID: PMC9248401 DOI: 10.1093/neuonc/noac022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The alternative lengthening of telomeres (ALT) pathway is essential for tumor proliferation in astrocytomas. The goal of this study was to identify metabolic alterations linked to the ALT pathway that can be exploited for noninvasive magnetic resonance spectroscopy (MRS)-based imaging of astrocytomas in vivo. METHODS Genetic and pharmacological methods were used to dissect the association between the ALT pathway and glucose metabolism in genetically engineered and patient-derived astrocytoma models. 2H-MRS was used for noninvasive imaging of ALT-linked modulation of glycolytic flux in mice bearing orthotopic astrocytomas in vivo. RESULTS The ALT pathway was associated with higher activity of the rate-limiting glycolytic enzyme phosphofructokinase-1 and concomitantly elevated flux of glucose to lactate in astrocytoma cells. Silencing the ALT pathway or treating with the poly(ADP-ribose) polymerase inhibitor niraparib that induces telomeric fusion in ALT-dependent astrocytoma cells abrogated glycolytic flux. Importantly, this metabolic reprogramming could be non-invasively visualized by 2H-MRS. Lactate production from [6,6'-2H]-glucose was higher in ALT-dependent astrocytoma tumors relative to the normal brain in vivo. Furthermore, treatment of orthotopic astrocytoma-bearing mice with niraparib reduced lactate production from [6,6'-2H]-glucose at early timepoints when alterations in tumor volume could not be detected by anatomical imaging, pointing to the ability of [6,6'-2H]-glucose to report on pseudoprogression in vivo. CONCLUSIONS We have mechanistically linked the ALT pathway to elevated glycolytic flux and demonstrated the ability of [6,6'-2H]-glucose to non-invasively assess tumor burden and response to therapy in astrocytomas. Our findings point to a novel, clinically translatable method for metabolic imaging of astrocytoma patients.
Collapse
Affiliation(s)
- Céline Taglang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Joydeep Mukherjee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Meryssa Tran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Donghyun Hong
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Hema Artee Luchman
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Russell O Pieper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Pavithra Viswanath
- Corresponding Author: Pavithra Viswanath, PhD, Department of Radiology and Biomedical Imaging, University of California, San Francisco, 1700 4th St, San Francisco, CA 94143, USA ()
| |
Collapse
|
11
|
Luchini C, Lawlor RT, Bersani S, Vicentini C, Paolino G, Mattiolo P, Pea A, Cingarlini S, Milella M, Scarpa A. Alternative Lengthening of Telomeres (ALT) in Pancreatic Neuroendocrine Tumors: Ready for Prime-Time in Clinical Practice? Curr Oncol Rep 2021; 23:106. [PMID: 34269919 PMCID: PMC8285324 DOI: 10.1007/s11912-021-01096-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 10/26/2022]
Abstract
PURPOSE OF REVIEW Alternative lengthening of telomeres (ALT) is a telomerase-independent mechanism used by some types of malignancies, including pancreatic neuroendocrine tumors, to overcome the issue of telomere shortening, thus supporting tumor growth and cell proliferation. This review is focused on the most important achievements and opportunities deriving from ALT assessment in PanNET onco-pathology, highlighting the most promising fields in which such biomarker could be implemented in clinical practice. RECENT FINDINGS In pancreatic neuroendocrine tumors (PanNET), ALT is strongly correlated with the mutational status of two chromatin remodeling genes, DAXX and ATRX. Recent advances in tumor biology permitted to uncover important roles of ALT in the landscape of PanNET, potentially relevant for introducing this biomarker into clinical practice. Indeed, ALT emerged as a reliable indicator of worse prognosis for PanNET, helping in clinical stratification and identification of "high-risk" patients. Furthermore, it is a very specific marker supporting the pancreatic origin of neuroendocrine neoplasms and can be used for improving the diagnostic workflow of patients presenting with neuroendocrine metastasis from unknown primary. The activation of this process can be determined by specific FISH analysis. ALT should be introduced in clinical practice for identifying "high-risk" PanNET patients and improving their clinical management, and as a marker of pancreatic origin among neuroendocrine tumors.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Samantha Bersani
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Caterina Vicentini
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Gaetano Paolino
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Antonio Pea
- Department of Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Sara Cingarlini
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Michele Milella
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134, Verona, Italy. .,ARC-Net Research Centre, University and Hospital Trust of Verona, 37134, Verona, Italy.
| |
Collapse
|
12
|
MacKenzie D, Watters AK, To JT, Young MW, Muratori J, Wilkoff MH, Abraham RG, Plummer MM, Zhang D. ALT Positivity in Human Cancers: Prevalence and Clinical Insights. Cancers (Basel) 2021; 13:2384. [PMID: 34069193 PMCID: PMC8156225 DOI: 10.3390/cancers13102384] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023] Open
Abstract
Many exciting advances in cancer-related telomere biology have been made in the past decade. Of these recent advances, great progress has also been made with respect to the Alternative Lengthening of Telomeres (ALT) pathway. Along with a better understanding of the molecular mechanism of this unique telomere maintenance pathway, many studies have also evaluated ALT activity in various cancer subtypes. We first briefly review and assess a variety of commonly used ALT biomarkers. Then, we provide both an update on ALT-positive (ALT+) tumor prevalence as well as a systematic clinical assessment of the presently studied ALT+ malignancies. Additionally, we discuss the pathogenetic alterations in ALT+ cancers, for example, the mutation status of ATRX and DAXX, and their correlations with the activation of the ALT pathway. Finally, we highlight important ALT+ clinical associations within each cancer subtype and subdivisions within, as well as their prognoses. We hope this alternative perspective will allow scientists, clinicians, and drug developers to have greater insight into the ALT cancers so that together, we may develop more efficacious treatments and improved management strategies to meet the urgent needs of cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Maria M. Plummer
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA; (D.M.J.); (A.K.W.); (J.T.T.); (M.W.Y.); (J.M.); (M.H.W.); (R.G.A.)
| | - Dong Zhang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA; (D.M.J.); (A.K.W.); (J.T.T.); (M.W.Y.); (J.M.); (M.H.W.); (R.G.A.)
| |
Collapse
|
13
|
Viswanath P, Batsios G, Ayyappan V, Taglang C, Gillespie AM, Larson PEZ, Luchman HA, Costello JF, Pieper RO, Ronen SM. Metabolic imaging detects elevated glucose flux through the pentose phosphate pathway associated with TERT expression in low-grade gliomas. Neuro Oncol 2021; 23:1509-1522. [PMID: 33864084 DOI: 10.1093/neuonc/noab093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Telomerase reverse transcriptase (TERT) is essential for tumor proliferation, including in low-grade oligodendrogliomas (LGOGs). Since TERT is silenced in normal cells, it is also a therapeutic target. Therefore, non-invasive methods of imaging TERT are needed. Here, we examined the link between TERT expression and metabolism in LGOGs, with the goal of leveraging this information for non-invasive magnetic resonance spectroscopy (MRS)-based metabolic imaging of LGOGs. METHODS Immortalized normal human astrocytes with doxycycline-inducible TERT silencing, patient-derived LGOG cells, orthotopic tumors and LGOG patient biopsies were studied to determine the mechanistic link between TERT expression and glucose metabolism. The ability of hyperpolarized [U- 13C, U- 2H]-glucose to non-invasively assess TERT expression was tested in live cells and orthotopic tumors. RESULTS TERT expression was associated with elevated glucose flux through the pentose phosphate pathway (PPP), elevated NADPH, which is a major product of the PPP, and elevated GSH, which is maintained in a reduced state by NADPH. Importantly, hyperpolarized [U- 13C, U- 2H]-glucose metabolism via the PPP non-invasively reported on TERT expression and response to TERT inhibition in patient-derived LGOG cells and orthotopic tumors. Mechanistically, TERT acted via the sirtuin SIRT2 to upregulate the glucose transporter GLUT1 and the rate-limiting PPP enzyme glucose-6-phosphate dehydrogenase. CONCLUSIONS We have, for the first time, leveraged a mechanistic understanding of TERT-associated metabolic reprogramming for non-invasive imaging of LGOGs using hyperpolarized [U- 13C, U- 2H]-glucose. Our findings provide a novel way of imaging a hallmark of tumor immortality and have the potential to improve diagnosis and treatment response assessment for LGOG patients.
Collapse
Affiliation(s)
- Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Vinay Ayyappan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Celiné Taglang
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - H Artee Luchman
- Department of Cell Biology and Anatomy and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Joseph F Costello
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Russell O Pieper
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Gao Y, Wu Y, Zhang N, Yuan H, Wang F, Xu H, Yu J, Ma J, Hou S, Cao X. IDH1 gene mutation activates Smad signaling molecules to regulate the expression levels of cell cycle and biological rhythm genes in human glioma U87‑MG cells. Mol Med Rep 2021; 23:354. [PMID: 33760141 PMCID: PMC7974315 DOI: 10.3892/mmr.2021.11993] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Isocitrate dehydrogenase1 (IDH1) mutation is the most important genetic change in glioma. The most common IDH1 mutation results in the amino acid substitution of arginine 132 (Arg/R132), which is located at the active site of the enzyme. IDH1 Arg132His (R132H) mutation can reduce the proliferative rate of glioma cells. Numerous diseases follow circadian rhythms, and there is growing evidence that circadian disruption may be a risk factor for cancer in humans. Dysregulation of the circadian clock serves an important role in the development of malignant tumors, including glioma. Brain-Muscle Arnt-Like protein 1 (BMAL1) and Circadian Locomotor Output Cycles Kaput (CLOCK) are the main biological rhythm genes. The present study aimed to further study whether there is an association between IDH1 R132H mutation and biological rhythm in glioma, and whether this affects the occurrence of glioma. The Cancer Genome Atlas (TCGA) database was used to detect the expression levels of the biological rhythm genes BMAL1 and CLOCK in various types of tumor. Additionally, U87-MG cells were infected with wild-type and mutant IDH1 lentiviruses. Colony formation experiments were used to detect cell proliferation in each group, cell cycle distribution was detected by flow cytometry and western blotting was used to detect the expression levels of wild-type and mutant IDH1, cyclins, biological rhythm genes and Smad signaling pathway-associated genes in U87-MG cells. TCGA database results suggested that BMAL1 and CLOCK were abnormally expressed in glioma. Cells were successfully infected with wild-type and mutant IDH1 lentiviruses. Colony formation assay revealed decreased cell proliferation in the IDH1 R132H mutant group. The cell cycle distribution detected by flow cytometry indicated that IDH1 gene mutation increased the G1 phase ratio and decreased the S phase ratio in U87-MG cells. The western blotting results demonstrated that IDH1 R132H mutation decreased the expression levels of the S phase-associated proteins Cyclin A and CDK2, and increased the expression levels of the G1 phase-associated proteins Cyclin D3 and CDK4, but did not significantly change the expression levels of the G2/M phase-associated protein Cyclin B1. The expression levels of the positive and negative rhythm regulation genes BMAL1, CLOCK, period (PER s (PER1, 2 and 3) and cryptochrom (CRY)s (CRY1 and 2) were significantly decreased, those of the Smad signaling pathway-associated genes Smad2, Smad3 and Smad2-3 were decreased, and those of phosphorylated (p)-Smad2, p-Smad3 and Smad4 were increased. Therefore, the present results suggested that the IDH1 R132H mutation may alter the cell cycle and biological rhythm genes in U87-MG cells through the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Yongying Gao
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yanwei Wu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ningmei Zhang
- Department of Pathology, Tumor Hospital, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hongmei Yuan
- Functional Department, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia 750021, P.R. China
| | - Fei Wang
- Department of Pathology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia 750001, P.R. China
| | - Hui Xu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiaxiang Yu
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jie Ma
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiangmei Cao
- Department of Pathology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
15
|
Powter B, Jeffreys SA, Sareen H, Cooper A, Brungs D, Po J, Roberts T, Koh ES, Scott KF, Sajinovic M, Vessey JY, de Souza P, Becker TM. Human TERT promoter mutations as a prognostic biomarker in glioma. J Cancer Res Clin Oncol 2021; 147:1007-1017. [PMID: 33547950 PMCID: PMC7954705 DOI: 10.1007/s00432-021-03536-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/15/2021] [Indexed: 12/27/2022]
Abstract
The TERT promoter (pTERT) mutations, C228T and C250T, play a significant role in malignant transformation by telomerase activation, oncogenesis and immortalisation of cells. C228T and C250T are emerging as important biomarkers in many cancers including glioblastoma multiforme (GBM), where the prevalence of these mutations is as high as 80%. Additionally, the rs2853669 single nucleotide polymorphism (SNP) may cooperate with these pTERT mutations in modulating progression and overall survival in GBM. Using liquid biopsies, pTERT mutations, C228T and C250T, and other clinically relevant biomarkers can be easily detected with high precision and sensitivity, facilitating longitudinal analysis throughout therapy and aid in cancer patient management. In this review, we explore the potential for pTERT mutation analysis, via liquid biopsy, for its potential use in personalised cancer therapy. We evaluate the relationship between pTERT mutations and other biomarkers as well as their potential clinical utility in early detection, prognostication, monitoring of cancer progress, with the main focus being on brain cancer.
Collapse
Affiliation(s)
- Branka Powter
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.
| | - Sarah A Jeffreys
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Heena Sareen
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,Western Clinical School, University of New South Wales South, Goulburn St, Liverpool, NSW, 2170, Australia
| | - Adam Cooper
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.,Cancer Therapy Centre, Liverpool Hospital, Elizabeth St and Goulburn St, Liverpool, NSW, 2170, Australia
| | - Daniel Brungs
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Joseph Po
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia
| | - Tara Roberts
- School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.,Western Clinical School, University of New South Wales South, Goulburn St, Liverpool, NSW, 2170, Australia
| | - Eng-Siew Koh
- Western Clinical School, University of New South Wales South, Goulburn St, Liverpool, NSW, 2170, Australia.,Cancer Therapy Centre, Liverpool Hospital, Elizabeth St and Goulburn St, Liverpool, NSW, 2170, Australia
| | - Kieran F Scott
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Mila Sajinovic
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia
| | - Joey Y Vessey
- Cancer Therapy Centre, Liverpool Hospital, Elizabeth St and Goulburn St, Liverpool, NSW, 2170, Australia
| | - Paul de Souza
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.,Western Clinical School, University of New South Wales South, Goulburn St, Liverpool, NSW, 2170, Australia.,Cancer Therapy Centre, Liverpool Hospital, Elizabeth St and Goulburn St, Liverpool, NSW, 2170, Australia.,School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Therese M Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.,Western Clinical School, University of New South Wales South, Goulburn St, Liverpool, NSW, 2170, Australia
| |
Collapse
|
16
|
Viswanath P, Batsios G, Mukherjee J, Gillespie AM, Larson PEZ, Luchman HA, Phillips JJ, Costello JF, Pieper RO, Ronen SM. Non-invasive assessment of telomere maintenance mechanisms in brain tumors. Nat Commun 2021; 12:92. [PMID: 33397920 PMCID: PMC7782549 DOI: 10.1038/s41467-020-20312-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/27/2020] [Indexed: 01/29/2023] Open
Abstract
Telomere maintenance is a universal hallmark of cancer. Most tumors including low-grade oligodendrogliomas use telomerase reverse transcriptase (TERT) expression for telomere maintenance while astrocytomas use the alternative lengthening of telomeres (ALT) pathway. Although TERT and ALT are hallmarks of tumor proliferation and attractive therapeutic targets, translational methods of imaging TERT and ALT are lacking. Here we show that TERT and ALT are associated with unique 1H-magnetic resonance spectroscopy (MRS)-detectable metabolic signatures in genetically-engineered and patient-derived glioma models and patient biopsies. Importantly, we have leveraged this information to mechanistically validate hyperpolarized [1-13C]-alanine flux to pyruvate as an imaging biomarker of ALT status and hyperpolarized [1-13C]-alanine flux to lactate as an imaging biomarker of TERT status in low-grade gliomas. Collectively, we have identified metabolic biomarkers of TERT and ALT status that provide a way of integrating critical oncogenic information into non-invasive imaging modalities that can improve tumor diagnosis and treatment response monitoring.
Collapse
Affiliation(s)
- Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Joydeep Mukherjee
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - H Artee Luchman
- Department of Cell Biology and Anatomy, Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Joanna J Phillips
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Joseph F Costello
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Russell O Pieper
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Correlation between IDH, ATRX, and TERT promoter mutations in glioma. Brain Tumor Pathol 2020; 37:33-40. [PMID: 32227259 DOI: 10.1007/s10014-020-00360-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
According to the 2016 World Health Organization (WHO) classification of central nervous system tumors, diffuse astrocytic and oligodendroglial tumors are differentiated by the presence of isocitrate dehydrogenase 1 or 2 (IDH1/2) mutation and the combined loss of the short arm of chromosome 1 and the long arm of chromosome 19 (1p/19q co-deletion). IDH-mutant astrocytoma often has p53 and alpha-thalassemia/mental retardation syndrome X-linked (ATRX) mutation, showing the alternative lengthening of telomeres (ALT) phenotype, while IDH-mutant and 1p/19q-co-deleted oligodendroglioma often have wild-type p53 and telomerase reverse transcriptase (TERT) promoter mutation, showing telomerase activation. This study analyzed IDH, ATRX, and TERT promoter mutations, and the correlation between them. Immortalized cells overcome the telomere-related crisis by activating telomerase or ALT. In glioma, telomerase is mainly activated by TERT promoter mutation, while ALT is usually associated with ATRX mutation. Although the mechanism of how ATRX mutation induces ALT remains unclear, ATRX loss alone is believed to be insufficient to induce ALT. Treatments targeting telomere maintenance are promising.
Collapse
|