1
|
Facundo AN, Magalhães M, Nascimento GC, Azulay RS, Santos RM, Freitas LA, Nascimento AGPAC, Rodrigues VP, Santos WC, Beckman AMGS, Abreu JMF, Silva RP, Carneiro EL, Oliveira Neto CP, Gil da Costa RM, Corcoy R, Mato E, Faria MS. The expression of VDACs and Bcl2 family genes in pituitary adenomas: clinical correlations and postsurgical outcomes. Front Endocrinol (Lausanne) 2024; 15:1481050. [PMID: 39449743 PMCID: PMC11499145 DOI: 10.3389/fendo.2024.1481050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/13/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Pituitary adenomas (PAs) are benign tumors with high prevalence and, occasionally, aggressive course. The tumorigenesis of these lesions is not completely understood at the molecular level. BAK1 and BAX proteins play fundamental roles in apoptosis and seem to interact with VDAC proteins, whose expressions have been markedly altered in cancer, impacting their prognosis. Objective to evaluate the gene expression of VDAC1, VDAC2, BAK1 and BAX and their association with clinical and imaging characteristics in PA. Methods Clinical-epidemiological data were collected from 117 tumor samples from patients affected by PA. Invasiveness was assessed by the Knosp scale. Gene expression was examined by real-time PCR. Relative expression analysis was performed by 2^(-DDCt) method. Results The sample was mainly composed of women (69/117 - 57.2%). Tumor subtypes observed were Non-Functioning (NF) (73/117 - 62.4%), Acromegaly (24/117 - 20.5%) and Cushing's Disease (CD) (20/117 - 17.1%). Compared to normal tissue, there was a significant reduction in VDAC1 expression in the Acromegaly (p=0.029) and NF (p=0.002) groups. BAX expression was lower in all groups (p <0.001; p=0.007; P =0.005). No difference was found in VDAC2 and BAK1 expression, compared to normal pituitary. Overexpression of VDAC2 occurred in PAs with post-surgical regrowth (p=0.042). A strongly negative correlation was observed in BAX and BAK1 expression in CD. Conclusion The results indicated that downregulations of VDAC1 and BAX may be related to resistance to apoptosis. In contrast, overexpression of VDAC2 in regrowing PAs suggests an antiapoptotic role for this gene. In summary, the genes evaluated might be involved in the biopathology of PAs.
Collapse
Affiliation(s)
- AN Facundo
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - M Magalhães
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - GC Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RS Azulay
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RM Santos
- Service of Radiology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - LA Freitas
- Service of Radiology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - AGPAC Nascimento
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Pathology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - VP Rodrigues
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Department of Morphology, Federal University of Maranhao (UFMA), São Luis, Brazil
| | - WC Santos
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - AMGS Beckman
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - JMF Abreu
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RP Silva
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - EL Carneiro
- Service of Neurosurgery, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - CP Oliveira Neto
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RM Gil da Costa
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Department of Morphology, Federal University of Maranhao (UFMA), São Luis, Brazil
| | - R Corcoy
- CIBER Bioengineering, Biomaterials and Nanotechnology (CIBER-BBN), Instituto de Salud III, Madrid, Spain
- Department of Nutricion and Endocrinology of Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - E Mato
- CIBER Bioengineering, Biomaterials and Nanotechnology (CIBER-BBN), Instituto de Salud III, Madrid, Spain
- Department of Nutricion and Endocrinology of Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - MS Faria
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| |
Collapse
|
2
|
Sablin MP, Gestraud P, Jonas SF, Lamy C, Lacroix-Triki M, Bachelot T, Filleron T, Lacroix L, Tran-Dien A, Jézéquel P, Mauduit M, Barros Monteiro J, Jimenez M, Michiels S, Attignon V, Soubeyran I, Driouch K, Servant N, Le Tourneau C, Kamal M, André F, Bièche I. Copy number alterations in metastatic and early breast tumours: prognostic and acquired biomarkers of resistance to CDK4/6 inhibitors. Br J Cancer 2024; 131:1060-1067. [PMID: 39090361 PMCID: PMC11405515 DOI: 10.1038/s41416-024-02804-6] [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: 02/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Copy number alterations (CNA) are acquired during the evolution of cancers from their early stage to metastatic stage. This study aims at analysing the clinical value of the identified metastasis-associated CNAs both in metastatic breast cancers (mBCs) and early breast cancers (eBCs). METHODS Single-nucleotide polymorphism (SNP)-array was performed on 926 biopsies from mBC patients, enrolled in SAFIR02-BREAST prospective trial. CNA profiles of eBCs from The Cancer Genome Atlas Breast Invasive Carcinoma (n = 770), Molecular Taxonomy of Breast Cancer International Consortium (n = 1620) and PACS04 trial (n = 243) cohorts were used as references for comparing mBCs and eBCs CNA profiles. Overall survival was the considered survival endpoint. RESULTS Among the twenty-one genes frequently altered in ER + /HER2- mBCs: focal amplification of TERT was associated with poor outcome in the ER + /HER2- mBC population. Among the ER + /HER2- mBCs patients for whom CDK4/6 inhibitors information before biopsies collection was available: we identified seven genes on post-treatment biopsies, including the cyclin-dependent kinase 4 (CDK4), which was amplified in 9.8% of the ER + /HER2- mBCs pretreated population, as compared to 1.5% in the ER + /HER2- mBCs unpretreated population (P = 2.82E-04) as well as the 3 eBC populations. CDK4 amplification was associated with poor outcome in the ER + /HER2- eBCs. CONCLUSIONS This study provides insights into the biology of mBCs and identifies clinically useful genomic features for future improvement of breast cancer patient management.
Collapse
Affiliation(s)
- Marie-Paule Sablin
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Pierre Gestraud
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Sarah Flora Jonas
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Oncostat U1018, Inserm, Université Paris-Saclay, Équipe Labellisée Ligue Contre le Cancer, Villejuif, France
| | - Constance Lamy
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | | | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Thomas Filleron
- Department of Biostatistics, Institut Claudius-Regaud, IUCT Oncopole, Toulouse, France
| | - Ludovic Lacroix
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
- Genomic Platform and Biobank, CNRS UMS3655-INSERM US23, AMMICA, Gustave Roussy, Villejuif, France
| | | | - Pascal Jézéquel
- Omics Data Science Unit, Institut de Cancérologie de l'Ouest (ICO), Angers-Nantes, France
| | | | | | | | - Stefan Michiels
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Oncostat U1018, Inserm, Université Paris-Saclay, Équipe Labellisée Ligue Contre le Cancer, Villejuif, France
| | | | | | - Keltouma Driouch
- Department of Genetics, Institut Curie, Paris University, Paris, France
| | - Nicolas Servant
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, Paris University, Paris, France.
| |
Collapse
|
3
|
Long W, Zeng YX, Zheng BX, Li YB, Wang YK, Chan KH, She MT, Lu YJ, Cao C, Wong WL. Targeting hTERT Promoter G-Quadruplex DNA Structures with Small-Molecule Ligand to Downregulate hTERT Expression for Triple-Negative Breast Cancer Therapy. J Med Chem 2024; 67:13363-13382. [PMID: 38987863 DOI: 10.1021/acs.jmedchem.4c01255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Human telomerase reverse transcriptase (hTERT) may have noncanonical functions in transcriptional regulation and metabolic reprogramming in cancer cells, but it is a challenging target. We thus developed small-molecule ligands targeting hTERT promoter G-quadruplex DNA structures (hTERT G4) to downregulate hTERT expression. Ligand 5 showed high affinity toward hTERT G4 (Kd = 1.1 μM) and potent activity against triple-negative breast cancer cells (MDA-MB-231, IC50 = 1 μM). In cell-based assays, 5 not only exerts markedly inhibitory activity on classical telomere functions including decreased telomerase activity, shortened telomere length, and cellular senescence but also induces DNA damage, acute cellular senescence, and apoptosis. This study reveals that hTERT G4-targeting ligand may cause mitochondrial dysfunction, disrupt iron metabolism and activate ferroptosis in cancer cells. The in vivo antitumor efficacy of 5 was also evaluated in an MDA-MB-231 xenograft mouse model and approximately 78.7% tumor weight reduction was achieved. No observable toxicity against the major organs was observed.
Collapse
Affiliation(s)
- Wei Long
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Yao-Xun Zeng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Bo-Xin Zheng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Yu-Bo Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Ya-Kun Wang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Ka-Hin Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Meng-Ting She
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology Guangzhou 510006, China
| | - Chunyang Cao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| |
Collapse
|
4
|
Staubitz-Vernazza JI, Müller C, Heymans A, Nedwed AS, Schindeldecker M, Hartmann M, Kloth M, Schad A, Roth W, Musholt TJ, Hartmann N. Gene Expression Profiles of AHNAK2, DCSTAMP, FN1, and TERT Correlate With Mutational Status and Recurrence in Papillary Thyroid Carcinoma. Genes Chromosomes Cancer 2024; 63:e23256. [PMID: 39193983 DOI: 10.1002/gcc.23256] [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: 02/23/2024] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 08/29/2024] Open
Abstract
Papillary thyroid carcinoma (PTC), the most common malignancy of follicular cell derivation, is generally associated with good prognosis. Nevertheless, it is important to identify patients with aggressive PTCs and unfavorable outcome. Molecular markers such as BRAFV600E mutation and TERT promoter mutations have been proposed for risk stratification. While TERT promoter mutations have been frequently associated with aggressive PTCs, the association of BRAFV600E mutation with increased recurrence and mortality is less clear and has been controversially discussed. The aim of the present study was to analyze whether differentially expressed genes can predict BRAFV600E mutations as well as TERT promoter mutations in PTCs. RNA sequencing identified a large number of differentially expressed genes between BRAFV600E and BRAFwildtype PTCs. Of those, AHNAK2, DCSTAMP, and FN1 could be confirmed in a larger cohort (n = 91) to be significantly upregulated in BRAFV600E mutant PTCs using quantitative RT-PCR. Moreover, individual PTC expression values of DCSTAMP and FN1 were able to predict the BRAFV600E mutation status with high sensitivity and specificity. The expression of TERT was detected in all PTCs harboring TERT promoter mutations and in 19% of PTCs without TERT promoter mutations. Tumors with both TERT expression and TERT promoter mutations were particularly associated with aggressive clinicopathological features and a shorter recurrence-free survival. Altogether, it will be interesting to explore the biological function of AHNAK2, DCSTAMP, and FN1 in PTC in more detail. The analysis of their expression patterns could allow the characterization of PTC subtypes and thus enabling a more individualized surgical and medical treatment.
Collapse
Affiliation(s)
- Julia I Staubitz-Vernazza
- Section of Endocrine Surgery, Department of General, Visceral and Transplantation Surgery, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Celine Müller
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Antonia Heymans
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Annekathrin Silvia Nedwed
- Institute for Medical Biometry, Epidemiology and Informatics, University Medical Centre Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Mario Schindeldecker
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Monika Hartmann
- Department of Medicine III, University Medical Centre Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Kloth
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Arno Schad
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas J Musholt
- Section of Endocrine Surgery, Department of General, Visceral and Transplantation Surgery, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nils Hartmann
- Institute of Pathology, University Medical Centre, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
5
|
Qiu C, Zhou Y, Xiao X, Song T, Zeng D, Peng J. Stratification of Lung Adenocarcinoma Patients Based on In Silico and Immunohistochemistry Analyses of Oxidative Stress-Related Genes. Cancer Biother Radiopharm 2024. [PMID: 38949986 DOI: 10.1089/cbr.2024.0094] [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: 07/03/2024] Open
Abstract
Background: Lung adenocarcinoma (LUAD) remains heterogeneous in the prognosis of patients; oxidative stress (OS) has been widely linked to cancer progression. Therefore, it is necessary to explore the prognostic value of the OS-associated genes in LUAD. Methods: An OS-associated prognostic signature was developed using the Cox regression and random forest model in The Cancer Genome Atlas-LUAD dataset. Kaplan-Meier (K-M) survival curve and time-dependent receiver operating characteristic (tROC) curves were applied to evaluate and validate the predictive accuracy of this signature among the training and testing cohorts. A nomogram was constructed and also verified by the concordance index (C-index), calibration curves, and tROC curves, respectively. ESTIMATE algorithm and CIBERSORT algorithms were conducted to explore the signature's immune characteristics. Core target genes of the prognostic signature were identified in the protein-protein interaction network. Results: A six OS-associated prognostic gene signature (CDC25C, ERO1A, GRIA1, TERT, CAV1, BDNF) was developed. The tROC and K-M survival curves in the training and testing cohorts revealed that the signature had good and robust predictive capability to predict the overall survival of LUAD patients. Meanwhile, the risk score was an independent prognostic factor influencing patients' overall survival. The results of the C-index (0.714), calibration curves, and the 1-, 2-, and 3-year tROC curves (area under the curve = 0.703, 0.737, and 0.723, respectively) suggested that the nomogram had good predictive efficacy and prognostic value for LUAD. Then, the authors found that the high-risk group may be depletion or loss of antitumor function of immune cells. Finally, 10 core genes of the signature were predicted. Conclusion: Their study may provide a novel understanding for the identification of prognostic stratification in LUAD patients, as well as the regulation of OS-associated genes in LUAD progression.
Collapse
Affiliation(s)
- Chongrong Qiu
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| | - Yuming Zhou
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| | - Xiaoliu Xiao
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| | - Tianjun Song
- Department of Medicine II, University Hospital, Munich, Germany
| | - Dongyun Zeng
- Clinicopathological Diagnosis & Research Center, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Jingliang Peng
- Department of Emergency, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| |
Collapse
|
6
|
Liu B, Liu XY, Wang GP, Chen YX. The immune cell infiltration-associated molecular subtypes and gene signature predict prognosis for osteosarcoma patients. Sci Rep 2024; 14:5184. [PMID: 38431660 PMCID: PMC10908810 DOI: 10.1038/s41598-024-55890-0] [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: 05/26/2023] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
Host immune dysregulation involves in the initiation and development of osteosarcoma (OS). However, the exact role of immune cells in OS remains unknown. We aimed to distinguish the molecular subtypes and establish a prognostic model in OS patients based on immunocyte infiltration. The gene expression profile and corresponding clinical feature of OS patients were obtained from TARGET and GSE21257 datasets. MCP-counter and univariate Cox regression analyses were applied to identify immune cell infiltration-related molecular subgroups. Functional enrichment analysis and immunocyte infiltration analysis were performed between two subgroups. Furthermore, Cox regression and LASSO analyses were performed to establish the prognostic model for the prediction of prognosis and metastasis in OS patients. The subgroup with low infiltration of monocytic lineage (ML) was related to bad prognosis in OS patients. 435 DEGs were screened between the two subgroups. Functional enrichment analysis revealed these DEGs were involved in immune- and inflammation-related pathways. Three important genes (including TERT, CCDC26, and IL2RA) were identified to establish the prognostic model. The risk model had good prognostic performance for the prediction of metastasis and overall survival in OS patients. A novel stratification system was established based on ML-related signature. The risk model could predict the metastasis and prognosis in OS patients. Our findings offered a novel sight for the prognosis and development of OS.
Collapse
Affiliation(s)
- Bin Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Xiang-Yang Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Guo-Ping Wang
- Department of Spine Surgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Yi-Xin Chen
- Department of Rehabilitation Medicine, Xiangya Hospital of Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
7
|
Engin AB, Engin A. Obesity-Senescence-Breast Cancer: Clinical Presentation of a Common Unfortunate Cycle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:821-850. [PMID: 39287873 DOI: 10.1007/978-3-031-63657-8_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
There are few convincing studies establishing the relationship between endogenous factors that cause obesity, cellular aging, and telomere shortening. Without a functional telomerase, a cell undergoing cell division has progressive telomere shortening. While obesity influences health and longevity as well as telomere dynamics, cellular senescence is one of the major drivers of the aging process and of age-related disorders. Oxidative stress induces telomere shortening, while decreasing telomerase activity. When progressive shortening of telomere length reaches a critical point, it triggers cell cycle arrest leading to senescence or apoptotic cell death. Telomerase activity cannot be detected in normal breast tissue. By contrast, maintenance of telomere length as a function of human telomerase is crucial for the survival of breast cancer cells and invasion. Approximately three-quarters of breast cancers in the general population are hormone-dependent and overexpression of estrogen receptors is crucial for their continued growth. In obesity, increasing leptin levels enhance aromatase messenger ribonucleic acid (mRNA) expression, aromatase content, and its enzymatic activity on breast cancer cells, simultaneously activating telomerase in a dose-dependent manner. Meanwhile, applied anti-estrogen therapy increases serum leptin levels and thus enhances leptin resistance in obese postmenopausal breast cancer patients. Many studies revealed that shorter telomeres of postmenopausal breast cancer have higher local recurrence rates and higher tumor grade. In this review, interlinked molecular mechanisms are looked over between the telomere length, lipotoxicity/glycolipotoxicity, and cellular senescence in the context of estrogen receptor alpha-positive (ERα+) postmenopausal breast cancers in obese women. Furthermore, the effect of the potential drugs, which are used for direct inhibition of telomerase and the inhibition of human telomerase reverse transcriptase (hTERT) or human telomerase RNA promoters as well as approved adjuvant endocrine therapies, the selective estrogen receptor modulator and selective estrogen receptor down-regulators are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
8
|
Semenza GL. Mechanisms of Breast Cancer Stem Cell Specification and Self-Renewal Mediated by Hypoxia-Inducible Factor 1. Stem Cells Transl Med 2023; 12:783-790. [PMID: 37768037 PMCID: PMC10726407 DOI: 10.1093/stcltm/szad061] [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: 03/22/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
Many advanced human cancers contain regions of intratumoral hypoxia, with O2 gradients extending to anoxia. Hypoxia-inducible factors (HIFs) are activated in hypoxic cancer cells and drive metabolic reprogramming, vascularization, invasion, and metastasis. Hypoxia induces breast cancer stem cell (BCSC) specification by inducing the expression and/or activity of the pluripotency factors KLF4, NANOG, OCT4, and SOX2. Recent studies have identified HIF-1-dependent expression of PLXNB3, NARF, and TERT in hypoxic breast cancer cells. PLXNB3 binds to and activates the MET receptor tyrosine kinase, leading to activation of the SRC non-receptor tyrosine kinase and subsequently focal adhesion kinase, which promotes cancer cell migration and invasion. PLXNB3-MET-SRC signaling also activates STAT3, a transcription factor that mediates increased NANOG gene expression. Hypoxia-induced NARF binds to OCT4 and serves as a coactivator by stabilizing OCT4 binding to the KLF4, NANOG, and SOX2 genes and by stabilizing the interaction of OCT4 with KDM6A, a histone demethylase that erases repressive trimethylation of histone H3 at lysine 27, thereby increasing KLF4, NANOG, and SOX2 gene expression. In addition to increasing pluripotency factor expression by these mechanisms, HIF-1 directly activates expression of the TERT gene encoding telomerase, the enzyme required for maintenance of telomeres, which is required for the unlimited self-renewal of BCSCs. HIF-1 binds to the TERT gene and recruits NANOG, which serves as a coactivator by promoting the subsequent recruitment of USP9X, a deubiquitinase that inhibits HIF-1α degradation, and p300, a histone acetyltransferase that mediates acetylation of H3K27, which is required for transcriptional activation.
Collapse
Affiliation(s)
- Gregg L Semenza
- Armstrong Oxygen Biology Research Center, Institute for Cell Engineering, and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Webersinke G, Burghofer J, Malli T, Rammer M, Jahn SW, Niendorf A, Tavassoli FA, Moinfar F. TERT Promoter Mutation c.-124C>T Commonly Occurs in Low-Grade Fibromatosis-like Metaplastic Breast Carcinoma. Arch Pathol Lab Med 2023; 147:1451-1457. [PMID: 36897999 DOI: 10.5858/arpa.2022-0159-oa] [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: 11/28/2022] [Indexed: 03/12/2023]
Abstract
CONTEXT.— Low-grade fibromatosis-like metaplastic carcinoma (FLMC) is a very rare subtype of triple-negative metaplastic (spindle cell) breast carcinoma. It is characterized by the proliferation of spindle cells closely resembling fibromatosis, which represents a benign fibroblastic/myofibroblastic breast proliferation. Unlike most triple-negative and basal-like breast cancers, FLMC has a very low potential for metastases, but demonstrates frequent local recurrences. OBJECTIVE.— To genetically characterize FLMC. DESIGN.— To this end, we analyzed 7 cases by targeted next-generation sequencing for 315 cancer-related genes and performed comparative microarray copy number analysis in 5 of these cases. RESULTS.— All cases shared TERT alterations (6 patients with recurrent c.-124C>T TERT promoter mutation and 1 patient with copy number gain encompassing the TERT locus), had oncogenic PIK3CA/PIK3R1 mutations (activation of the PI3K/AKT/mTOR pathway), and lacked mutations in TP53. TERT was overexpressed in all FLMCs. CDKN2A/B loss or mutation was observed in 4 of 7 cases (57%). Furthermore, tumors displayed chromosomal stability, with only few copy number variations and a low tumor mutational burden. CONCLUSIONS— We conclude that FLMCs typically show the recurrent TERT promoter mutation c.-124C>T, activation of the PI3K/AKT/mTOR pathway, low genomic instability, and wild-type TP53. In conjunction with previous data of metaplastic (spindle cell) carcinoma with and without fibromatosis-like morphology, FLMC is most likely distinguished by TERT promoter mutation. Thus, our data support the notion of a distinct subgroup within low-grade metaplastic breast cancer with spindle cell morphology and associated TERT mutations.
Collapse
Affiliation(s)
- Gerald Webersinke
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Jonathan Burghofer
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Theodora Malli
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Melanie Rammer
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Stephan Wenzel Jahn
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria (Jahn, Moinfar)
| | - Axel Niendorf
- Department of Pathology and Molecular Diagnostic, Hamburg-West, Hamburg, Germany (Niendorf)
| | - Fattaneh A Tavassoli
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut (Tavassoli)
| | - Farid Moinfar
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria (Jahn, Moinfar)
- Department of Pathology and Molecular Pathology, Vinzenz Pathologieverbund Linz, Austria (Moinfar)
- The Vincent Academy of Pathology, Linz, Austria (Moinfar)
| |
Collapse
|
10
|
Tornesello ML, Cerasuolo A, Starita N, Amiranda S, Bonelli P, Tuccillo FM, Buonaguro FM, Buonaguro L, Tornesello AL. Reactivation of telomerase reverse transcriptase expression in cancer: the role of TERT promoter mutations. Front Cell Dev Biol 2023; 11:1286683. [PMID: 38033865 PMCID: PMC10684755 DOI: 10.3389/fcell.2023.1286683] [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: 08/31/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Telomerase activity and telomere elongation are essential conditions for the unlimited proliferation of neoplastic cells. Point mutations in the core promoter region of the telomerase reverse transcriptase (TERT) gene have been found to occur at high frequencies in several tumour types and considered a primary cause of telomerase reactivation in cancer cells. These mutations promote TERT gene expression by multiple mechanisms, including the generation of novel binding sites for nuclear transcription factors, displacement of negative regulators from DNA G-quadruplexes, recruitment of epigenetic activators and disruption of long-range interactions between TERT locus and telomeres. Furthermore, TERT promoter mutations cooperate with TPP1 promoter nucleotide changes to lengthen telomeres and with mutated BRAF and FGFR3 oncoproteins to enhance oncogenic signalling in cancer cells. TERT promoter mutations have been recognized as an early marker of tumour development or a major indicator of poor outcome and reduced patients survival in several cancer types. In this review, we summarize recent findings on the role of TERT promoter mutations, telomerase expression and telomeres elongation in cancer development, their clinical significance and therapeutic opportunities.
Collapse
Affiliation(s)
- Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Andrea Cerasuolo
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Noemy Starita
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Sara Amiranda
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Patrizia Bonelli
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Franca Maria Tuccillo
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Anna Lucia Tornesello
- Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
11
|
Lyu SI, Popp FC, Simon AG, Schultheis AM, Zander T, Fretter C, Schröder W, Bruns CJ, Schmidt T, Quaas A, Knipper K. Copy-number-gain of telomerase reverse transcriptase (hTERT) is associated with an unfavorable prognosis in esophageal adenocarcinoma. Sci Rep 2023; 13:17699. [PMID: 37848472 PMCID: PMC10582081 DOI: 10.1038/s41598-023-44844-7] [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: 08/27/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023] Open
Abstract
Esophageal adenocarcinoma exhibits one of the highest mortality rates among all cancer entities. Multimodal therapy strategies have improved patients' survival significantly. However, patients in early stages are currently limited to receiving only local therapies, even though some patients within this group showcase short survival periods. Until now, there has been no widely established clinically used biomarker to detect these high-risk patients. Telomerase reverse transcriptase (TERT), a gene encoding a crucial subunit of the telomerase enzyme, plays a significant role in establishing cancer cell immortality and is under suspicion for its potential contribution to tumor progression. Therefore, we aimed to evaluate the clinical relevance of the TERT amplification status. We included 643 patients with esophageal adenocarcinoma, who underwent Ivor-Lewis esophagectomy at the University Hospital of Cologne. The TERT amplification status was characterized using fluorescence in situ hybridization. Clinicopathological values and patients' overall survival were compared between patients with and without TERT amplification. Further sub-cohort analyses were conducted for patients with pT1N0-3 tumor stage. Eighty-One patients (12.6%) exhibited TERT amplification. Patients with amplified TERT showed significantly worse overall survival (median OS: 22.6 vs. 36.8 months, p = 0.009). Interestingly, TERT amplification could be characterized as an independent risk factor for worse overall survival in multivariate analysis in patients with pT1N0-3 tumor stage (HR = 2.440, 95% CI 1.095-5.440, p = 0.029). In this study, we describe the TERT amplification status as an independent risk factor for worse survival in patients diagnosed with esophageal adenocarcinoma at pT1N0-3 tumor stage, encompassing cases involving tumor infiltration of the lamina propria, muscularis mucosae, and/or submucosa. Based on our findings, we put forth the proposition that evaluating the TERT amplification status may serve as a valuable tool in identifying a specific subgroup of patients, namely those with TERT amplification and pT1N0-3 tumor-stage esophageal adenocarcinoma. The patients of this subgroup could potentially benefit from enhanced follow-up protocols, more aggressive treatment approaches, or possible targeted TERT inhibition therapies, all aimed at improving their overall clinical outcomes.
Collapse
Affiliation(s)
- Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Anne Maria Schultheis
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Faculty of Medicine and University Hospital of Cologne, Department of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
| | - Caroline Fretter
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.
| |
Collapse
|
12
|
Lei T, Yang Y, Shi Y, Deng X, Peng Y, Wang H, Chen T. Clinicopathological features and genomic profiles of a group of secretory breast carcinomas in which progressive cases have more complex genomic features. Diagn Pathol 2022; 17:101. [PMID: 36585729 PMCID: PMC9805283 DOI: 10.1186/s13000-022-01284-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Secretory breast carcinoma (SBC) is a rare malignant breast neoplasm with distinct histological features, including solid, microcystic, tubular, and rarely papillary structures, traditionally characterized by a t (12;15) (p13:q25) translocation, which usually leads to ETV6-NTRK3 fusion, suggesting an early event in tumorigenesis. Due to the rarity of this disease, very few genome sequencing studies have been performed on a series of cases, especially progressive cases. METHODS Seven lesions from 5 patients diagnosed at the Third Affiliated Hospital of Soochow University from 2007 to 2021 were included. Clinicopathological features and prognosis/survival data were collected. Next-generation DNA sequencing was performed on six of the seven lesions. RESULTS In total, 3/7 (42.9%) lesions demonstrated estrogen receptor (ER) expression, including weak, moderate to strong staining, and no lesion demonstrated progesterone receptor (PR) expression. There were no cases of human epidermal growth factor (HER2) overexpression, and the Ki-67 index was low. S-100 and pan-TRK protein were diffusely positively expressed in all cases. All lesions were characterized by a t(12;15) (p13:q25) translocation, leading to ETV6-NTRK3 fusion confirmed by fluorescence in situ hybridization (FISH). The sequencing results showed that ETV6-NTRK3 fusion was the main driver of early tumorigenesis, while SBC with invasive biological behavior had more complex genomic variation in which TERT promoter mutation was detected. CONCLUSIONS Immunohistochemical staining of a biomarker panel, including ER, PR, HER2, Ki-67, S-100 and pan-TRK, can be used as an auxiliary diagnostic tool, and FISH detection can be used as a diagnostic tool. ETV6-NTRK3 gene fusion involving multiple sites may drive tumorigenesis, while mutations in the TERT promoter region may be a factor driving tumor progression.
Collapse
Affiliation(s)
- Ting Lei
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| | - Yuyan Yang
- grid.412648.d0000 0004 1798 6160Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, 300211 P.R. China
| | - Yongqiang Shi
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| | - Xu Deng
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| | - Yan Peng
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| | - Hui Wang
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| | - Tongbing Chen
- grid.452253.70000 0004 1804 524XDepartment of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 P.R. China
| |
Collapse
|
13
|
Apolónio JD, Dias JS, Fernandes MT, Komosa M, Lipman T, Zhang CH, Leão R, Lee D, Nunes NM, Maia AT, Morera JL, Vicioso L, Tabori U, Castelo-Branco P. THOR is a targetable epigenetic biomarker with clinical implications in breast cancer. Clin Epigenetics 2022; 14:178. [PMID: 36529814 PMCID: PMC9759897 DOI: 10.1186/s13148-022-01396-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequently diagnosed cancer and a leading cause of death among women worldwide. Early BC is potentially curable, but the mortality rates still observed among BC patients demonstrate the urgent need of novel and more effective diagnostic and therapeutic options. Limitless self-renewal is a hallmark of cancer, governed by telomere maintenance. In around 95% of BC cases, this process is achieved by telomerase reactivation through upregulation of the human telomerase reverse transcriptase (hTERT). The hypermethylation of a specific region within the hTERT promoter, termed TERT hypermethylated oncological region (THOR) has been associated with increased hTERT expression in cancer. However, its biological role and clinical potential in BC have never been studied to the best of our knowledge. Therefore, we aimed to investigate the role of THOR as a biomarker and explore the functional impact of THOR methylation status in hTERT upregulation in BC. RESULTS THOR methylation status in BC was assessed by pyrosequencing on discovery and validation cohorts. We found that THOR is significantly hypermethylated in malignant breast tissue when compared to benign tissue (40.23% vs. 12.81%, P < 0.0001), differentiating malignant tumor from normal tissue from the earliest stage of disease. Using a reporter assay, the addition of unmethylated THOR significantly reduced luciferase activity by an average 1.8-fold when compared to the hTERT core promoter alone (P < 0.01). To further investigate its biological impact on hTERT transcription, targeted THOR demethylation was performed using novel technology based on CRISPR-dCas9 system and significant THOR demethylation was achieved. Cells previously demethylated on THOR region did not develop a histologic cancer phenotype in in vivo assays. Additional studies are required to validate these observations and to unravel the causality between THOR hypermethylation and hTERT upregulation in BC. CONCLUSIONS THOR hypermethylation is an important epigenetic mark in breast tumorigenesis, representing a promising biomarker and therapeutic target in BC. We revealed that THOR acts as a repressive regulatory element of hTERT and that its hypermethylation is a relevant mechanism for hTERT upregulation in BC.
Collapse
Affiliation(s)
- Joana Dias Apolónio
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - João S Dias
- University Hospital Center of Algarve, Faro, Portugal
| | - Mónica Teotónio Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Escola Superior de Saúde (ESSUAlg), Universidade Do Algarve, Faro, Portugal
| | - Martin Komosa
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Tatiana Lipman
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Cindy H Zhang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ricardo Leão
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Donghyun Lee
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Nuno Miguel Nunes
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ana-Teresa Maia
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
- Center for Research in Health Technologies and Information Systems (CINTESIS@RISE), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal
| | - José L Morera
- University Hospital Center of Algarve, Faro, Portugal
| | - Luis Vicioso
- Faculty of Medicine, Department of Histology and Pathological Anatomy, University of Malaga, Malaga, Spain
| | - Uri Tabori
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Pedro Castelo-Branco
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Gambelas Campus, Bld. 2, 8005-139, Faro, Portugal.
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal.
- Algarve Biomedical Center (ABC), University of Algarve, Gambelas Campus, 8005-139, Faro, Portugal.
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
14
|
Standing S, Tran S, Murguia-Favela L, Kovalchuk O, Bose P, Narendran A. Identification of Altered Primary Immunodeficiency-Associated Genes and Their Implications in Pediatric Cancers. Cancers (Basel) 2022; 14:5942. [PMID: 36497424 PMCID: PMC9741011 DOI: 10.3390/cancers14235942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Cancer is the leading cause of disease-related mortality in children and malignancies are more frequently observed in individuals with primary immunodeficiencies (PIDs). This study aimed to identify and highlight the molecular mechanisms, such as oncogenesis and immune evasion, by which PID-related genes may lead to the development of pediatric cancers. METHOD We implemented a novel bioinformatics framework using patient data from the TARGET database and performed a comparative transcriptome analysis of PID-related genes in pediatric cancers between normal and cancer tissues, gene ontology enrichment, and protein-protein interaction analyses, and determined the prognostic impacts of commonly mutated and differentially expressed PID-related genes. RESULTS From the Fulgent Genetics Comprehensive Primary Immunodeficiency panel of 472 PID-related genes, 89 genes were significantly differentially expressed between normal and cancer tissues, and 20 genes were mutated in two or more patients. Enrichment analysis highlighted many immune system processes as well as additional pathways in the mutated PID-related genes related to oncogenesis. Survival outcomes for patients with altered PID-related genes were significantly different for 75 of the 89 DEGs, often resulting in a poorer prognosis. CONCLUSIONS Overall, multiple PID-related genes demonstrated the connection between PIDs and cancer development and should be studied further, with hopes of identifying new therapeutic targets.
Collapse
Affiliation(s)
- Shaelene Standing
- Section of Pediatric Oncology and Blood and Marrow Transplantation, Division of Pediatrics, Alberta Children’s Hospital and University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Son Tran
- Section of Pediatric Oncology and Blood and Marrow Transplantation, Division of Pediatrics, Alberta Children’s Hospital and University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Luis Murguia-Favela
- Section of Pediatric Hematology and Immunology, Division of Pediatrics, Alberta Children’s Hospital and University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Pinaki Bose
- Departments of Oncology, Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Aru Narendran
- Section of Pediatric Oncology and Blood and Marrow Transplantation, Division of Pediatrics, Alberta Children’s Hospital and University of Calgary, Calgary, AB T3B 6A8, Canada
| |
Collapse
|
15
|
Ott P, Araúzo-Bravo MJ, Hoffmann MJ, Poyet C, Bendhack ML, Santourlidis S, Erichsen L. Differential DNA Methylation of THOR and hTAPAS in the Regulation of hTERT and the Diagnosis of Cancer. Cancers (Basel) 2022; 14:cancers14184384. [PMID: 36139544 PMCID: PMC9497117 DOI: 10.3390/cancers14184384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Because of its high prevalence of >45% in 9 out of 11 (82%) cancer types screened, THOR hypermethylation has been suggested to be a frequent telomerase-activating mechanism in hTERT-expressing tumor types, e.g., in cancers of the prostate, breast, blood, colon, lung, bladder, and brain. In this prime example, we present detailed DNA methylation profiles in urothelial cancer that reveal the exact positions of the most differentially methylated CpG dinucleotides within the THOR region in order to design an efficient Methylation-Specific PCR (MSPCR) approach for diagnostic and prognostic purposes. Furthermore, our data suggest an epigenetic mechanism regulating hTERT expression through the methylation status of THOR and lncRNA hTAPAS. Abstract Background: Although DNA methylation in the gene promoters usually represses gene expression, the TERT hypermethylated oncological region (THOR) located 5′ of the hTERT gene is hypermethylated when hTERT is expressed in diverse cancer types, including urothelial cancer (UC). Methods: Comprehensive MeDIP and DNA methylation array analyses complemented by the technically independent method of bisulfite genomic sequencing were applied on pathologically reviewed and classified urothelial carcinoma specimens and healthy urothelial tissue samples to reveal the methylation status of THOR in detail. Results: The detailed DNA methylation profiles reveal the exact positions of differentially methylated CpG dinucleotides within THOR in urothelial cancer and provide evidence ofa diverging role of methylation of these CpGs in the regulation of hTERT. In particular, our data suggest a regulating mechanism in which THOR methylation acts on hTERT expression through epigenetic silencing of the lncRNA hTERT antisense promoter-associated (hTAPAS), which represses hTERT. Conclusions: These findings precisely define the most differentially methylated CpGs of THOR in early urothelial cancer, enabling optimal design of Methylation-Specific PCR (MSPCR) primers to reliably probe these methylation differences for diagnostic and prognostic purposes. In addition, this strategy presents a prime example that is also applicable to many other malignancies. Finally, the first evidence for the underlying epigenetic mechanism regulating hTERT expression through the methylation status of THOR is provided.
Collapse
Affiliation(s)
- Pauline Ott
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Marcos J. Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Michèle J. Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Cedric Poyet
- Department of Urology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Marcelo L. Bendhack
- Department of Urology, University Hospital, Positivo University, Curitiba 80420-011, Brazil
| | - Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
- Correspondence: (S.S.); (L.E.)
| | - Lars Erichsen
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
- Correspondence: (S.S.); (L.E.)
| |
Collapse
|
16
|
Aslam J, Ardanza-Trevijano S, Xiong J, Arsuaga J, Sazdanovic R. TAaCGH Suite for Detecting Cancer-Specific Copy Number Changes Using Topological Signatures. ENTROPY 2022; 24:e24070896. [PMID: 35885119 PMCID: PMC9318413 DOI: 10.3390/e24070896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/13/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
Copy number changes play an important role in the development of cancer and are commonly associated with changes in gene expression. Persistence curves, such as Betti curves, have been used to detect copy number changes; however, it is known these curves are unstable with respect to small perturbations in the data. We address the stability of lifespan and Betti curves by providing bounds on the distance between persistence curves of Vietoris–Rips filtrations built on data and slightly perturbed data in terms of the bottleneck distance. Next, we perform simulations to compare the predictive ability of Betti curves, lifespan curves (conditionally stable) and stable persistent landscapes to detect copy number aberrations. We use these methods to identify significant chromosome regions associated with the four major molecular subtypes of breast cancer: Luminal A, Luminal B, Basal and HER2 positive. Identified segments are then used as predictor variables to build machine learning models which classify patients as one of the four subtypes. We find that no single persistence curve outperforms the others and instead suggest a complementary approach using a suite of persistence curves. In this study, we identified new cytobands associated with three of the subtypes: 1q21.1-q25.2, 2p23.2-p16.3, 23q26.2-q28 with the Basal subtype, 8p22-p11.1 with Luminal B and 2q12.1-q21.1 and 5p14.3-p12 with Luminal A. These segments are validated by the TCGA BRCA cohort dataset except for those found for Luminal A.
Collapse
Affiliation(s)
- Jai Aslam
- Department of Mathematics, NC State University, Raleigh, NC 27695, USA;
| | - Sergio Ardanza-Trevijano
- Department of Physics and Applied Mathematics, University of Navarra, 31008 Pamplona, Spain;
- Institute for Data Science and Artificial Intelligence, University of Navarra, 31009 Pamplona, Spain
| | - Jingwei Xiong
- Graduate Group in Biostatistics University of California Davis, Davis, CA 95616, USA;
| | - Javier Arsuaga
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA
- Department of Mathematics, University of California Davis, Davis, CA 95616, USA
- Correspondence: (J.A.); (R.S.)
| | - Radmila Sazdanovic
- Department of Mathematics, NC State University, Raleigh, NC 27695, USA;
- Correspondence: (J.A.); (R.S.)
| |
Collapse
|
17
|
Dratwa M, Wysoczanska B, Brankiewicz W, Stachowicz-Suhs M, Wietrzyk J, Matkowski R, Ekiert M, Szelachowska J, Maciejczyk A, Szajewski M, Baginski M, Bogunia-Kubik K. Relationship between Telomere Length, TERT Genetic Variability and TERT, TP53, SP1, MYC Gene Co-Expression in the Clinicopathological Profile of Breast Cancer. Int J Mol Sci 2022; 23:5164. [PMID: 35563554 PMCID: PMC9102200 DOI: 10.3390/ijms23095164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/15/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
The molecular mechanisms of telomerase reverse transcriptase (TERT) upregulation in breast cancer (BC) are complex. We compared genetic variability within TERT and telomere length with the clinical data of patients with BC. Additionally, we assessed the expression of the TERT, MYC, TP53 and SP1 genes in BC patients and in BC organoids (3D cell cultures obtained from breast cancer tissues). We observed the same correlation in the blood of BC patients and in BC organoids between the expression of TERT and TP53. Only in BC patients was a correlation found between the expression of the TERT and MYC genes and between TP53 and MYC. We found associations between TERT genotypes (rs2735940 and rs10069690) and TP53 expression and telomere length. BC patients with the TT genotype rs2735940 have a shorter telomere length, but patients with A allele rs10069690 have a longer telomere length. BC patients with a short allele VNTR-MNS16A showed higher expression of the SP1 and had a longer telomere. Our results bring new insight into the regulation of TERT, MYC, TP53 and SP1 gene expression related to TERT genetic variability and telomere length. Our study also showed for the first time a similar relationship in the expression of the above genes in BC patients and in BC organoids. These findings suggest that TERT genetic variability, expression and telomere length might be useful biomarkers for BC, but their prognostic value may vary depending on the clinical parameters of BC patients and tumor aggressiveness.
Collapse
Affiliation(s)
- Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Barbara Wysoczanska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Wioletta Brankiewicz
- Department of Pharmaceutical Technology and Biochemistry Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (W.B.); (M.B.)
| | - Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.S.-S.); (J.W.)
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.S.-S.); (J.W.)
| | - Rafał Matkowski
- Breast Unit, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland; (R.M.); (M.E.); (J.S.); (A.M.)
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Marcin Ekiert
- Breast Unit, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland; (R.M.); (M.E.); (J.S.); (A.M.)
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Jolanta Szelachowska
- Breast Unit, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland; (R.M.); (M.E.); (J.S.); (A.M.)
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Adam Maciejczyk
- Breast Unit, Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413 Wroclaw, Poland; (R.M.); (M.E.); (J.S.); (A.M.)
- Department of Oncology, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Mariusz Szajewski
- Department of Oncological Surgery, Gdynia Oncology Centre, 81-519 Gdynia, Poland;
- Division of Propaedeutics of Oncology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Maciej Baginski
- Department of Pharmaceutical Technology and Biochemistry Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (W.B.); (M.B.)
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| |
Collapse
|
18
|
Diefenbach RJ, Lee JH, Stewart A, Menzies AM, Carlino MS, Saw RPM, Stretch JR, Long GV, Scolyer RA, Rizos H. Anchored Multiplex PCR Custom Melanoma Next Generation Sequencing Panel for Analysis of Circulating Tumor DNA. Front Oncol 2022; 12:820510. [PMID: 35494035 PMCID: PMC9039342 DOI: 10.3389/fonc.2022.820510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Detection of melanoma mutations using circulating tumor DNA (ctDNA) is a potential alternative to using genomic DNA from invasive tissue biopsies. To date, mutations in the GC-rich TERT promoter region, which is commonly mutated in melanoma, have been technically difficult to detect in ctDNA using next-generation sequencing (NGS) panels. In this study, we developed a custom melanoma NGS panel for detection of ctDNA, which encompasses the top 15 gene mutations in melanoma including the TERT promoter. We analyzed 21 stage III and IV melanoma patient samples who were treatment-naïve or on therapy. The overall detection rate of the custom panel, based on BRAF/NRAS/TERT promoter mutations, was 14/21 (67%) patient samples which included a TERT C250T mutation in one BRAF and NRAS mutation negative sample. A BRAF or NRAS mutation was detected in the ctDNA of 13/21 (62%) patients while TERT promoter mutations were detected in 10/21 (48%) patients. Co-occurrence of TERT promoter mutations with BRAF or NRAS mutations was found in 9/10 (90%) patients. The custom ctDNA panel showed a concordance of 16/21 (76%) with tissue based-detection and included 12 BRAF/NRAS mutation positive and 4 BRAF/NRAS mutation negative patients. The ctDNA mutation detection rate for stage IV was 12/16 (75%) and for stage III was 1/5 (20%). Based on BRAF, NRAS and TERT promoter mutations, the custom melanoma panel displayed a limit of detection of ~0.2% mutant allele frequency and showed significant correlation with droplet digital PCR. For one patient, a novel MAP2K1 H119Y mutation was detected in an NRAS/BRAF/TERT promoter mutation negative background. To increase the detection rate to >90% for stage IV melanoma patients, we plan to expand our custom panel to 50 genes. This study represents one of the first to successfully detect TERT promoter mutations in ctDNA from cutaneous melanoma patients using a targeted NGS panel.
Collapse
Affiliation(s)
- Russell J Diefenbach
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Jenny H Lee
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Ashleigh Stewart
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jonathan R Stretch
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
19
|
Lee DD, Komosa M, Sudhaman S, Leão R, Zhang CH, Apolonio JD, Hermanns T, Wild PJ, Klocker H, Nassiri F, Zadeh G, Diplas BH, Yan H, Gallinger S, Pugh TJ, Ramaswamy V, Taylor MD, Castelo-Branco P, Nunes NM, Tabori U. Dual role of allele-specific DNA hypermethylation within the TERT promoter in cancer. J Clin Invest 2021; 131:146915. [PMID: 34720085 DOI: 10.1172/jci146915] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Aberrant activation of telomerase in human cancer is achieved by various alterations within the TERT promoter, including cancer-specific DNA hypermethylation of the TERT hypermethylated oncological region (THOR). However, the impact of allele-specific DNA methylation within the TERT promoter on gene transcription remains incompletely understood. Using allele-specific next-generation sequencing, we screened a large cohort of normal and tumor tissues (n = 652) from 10 cancer types and identified that differential allelic methylation (DAM) of THOR is restricted to cancerous tissue and commonly observed in major cancer types. THOR-DAM was more common in adult cancers, which develop through multiple stages over time, than in childhood brain tumors. Furthermore, THOR-DAM was especially enriched in tumors harboring the activating TERT promoter mutations (TPMs). Functional studies revealed that allele-specific gene expression of TERT requires hypomethylation of the core promoter, both in TPM and TERT WT cancers. However, the expressing allele with hypomethylated core TERT promoter universally exhibits hypermethylation of THOR, while the nonexpressing alleles are either hypermethylated or hypomethylated throughout the promoter. Together, our findings suggest a dual role for allele-specific DNA methylation within the TERT promoter in the regulation of TERT expression in cancer.
Collapse
Affiliation(s)
- Donghyun D Lee
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Martin Komosa
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sumedha Sudhaman
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ricardo Leão
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cindy H Zhang
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joana D Apolonio
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas Hermanns
- Department of Urology, University Hospital Zürich, University of Zurich, Zurich, Switzerland
| | - Peter J Wild
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Germany.,Frankfurt Institute for Advanced Studies (FIAS), Frankfurt, Germany
| | - Helmut Klocker
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Farshad Nassiri
- Division of Neurosurgery, University of Toronto, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Division of Neurosurgery, University of Toronto, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Bill H Diplas
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Hai Yan
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Pedro Castelo-Branco
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal.,Algarve Biomedical Center Research Institute, Faro, Portugal.,Centre for Biomedical Research, University of Algarve, Faro, Portugal.,Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Nuno Miguel Nunes
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Uri Tabori
- Program in Genetics and Genome Biology and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Dlouhy I, Karube K, Enjuanes A, Salaverria I, Nadeu F, Ramis-Zaldivar JE, Valero JG, Rivas-Delgado A, Magnano L, Martin-García D, Pérez-Galán P, Clot G, Rovira J, Jares P, Balagué O, Giné E, Mozas P, Briones J, Sancho JM, Salar A, Mercadal S, Alcoceba M, Valera A, Campo E, López-Guillermo A. Revised International Prognostic Index and genetic alterations are associated with early failure to R-CHOP in patients with diffuse large B-cell lymphoma. Br J Haematol 2021; 196:589-598. [PMID: 34632572 DOI: 10.1111/bjh.17858] [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: 06/14/2021] [Revised: 08/26/2021] [Accepted: 09/12/2021] [Indexed: 11/28/2022]
Abstract
Relapsed or refractory diffuse large B-cell lymphoma (DLBCL) cases have a poor outcome. Here we analysed clinico-biological features in 373 DLBCL patients homogeneously treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP), in order to identify variables associated with early failure to treatment (EF), defined as primary refractoriness or relapse within 12 months from diagnosis. In addition to clinical features, mutational status of 106 genes was studied by targeted next-generation sequencing in 111 cases, copy number alterations in 87, and gene expression profile (GEP) in 39. Ninety-seven cases (26%) were identified as EF and showed significantly shorter overall survival (OS). Patients with B symptoms, advanced stage, high levels of serum lactate dehydrogenase (LDH) or β2-microglobulin, low lymphocyte/monocyte ratio and higher Revised International Prognostic Index (R-IPI) scores, as well as those with BCL2 rearrangements more frequently showed EF, with R-IPI being the most important in logistic regression. Mutations in NOTCH2, gains in 5p15·33 (TERT), 12q13 (CDK2), 12q14·1 (CDK4) and 12q15 (MDM2) showed predictive importance for EF independently from R-IPI. GEP studies showed that EF cases were significantly enriched in sets related to cell cycle regulation and inflammatory response, while cases in response showed over-representation of gene sets related to extra-cellular matrix and tumour microenvironment.
Collapse
Affiliation(s)
- Ivan Dlouhy
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Kennosuke Karube
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Cell Biology & Pathology Department, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Anna Enjuanes
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Itziar Salaverria
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ferran Nadeu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan Enric Ramis-Zaldivar
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Juan G Valero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alfredo Rivas-Delgado
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Laura Magnano
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - David Martin-García
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Pérez-Galán
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Guillem Clot
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordina Rovira
- Department of Hematology, Hospital Clínic, Barcelona, Spain
| | - Pedro Jares
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Olga Balagué
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eva Giné
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain
| | - Pablo Mozas
- Department of Hematology, Hospital Clínic, Barcelona, Spain
| | | | | | | | | | - Miguel Alcoceba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Hospital Clínico Universitario, Salamanca, Spain
| | - Alexandra Valera
- Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elías Campo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,Institut d`Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,University of Barcelona, Barcelona, Spain
| | - Armando López-Guillermo
- Department of Hematology, Hospital Clínic, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Tumores Hematológicos, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
HIF-1 recruits NANOG as a coactivator for TERT gene transcription in hypoxic breast cancer stem cells. Cell Rep 2021; 36:109757. [PMID: 34592152 DOI: 10.1016/j.celrep.2021.109757] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/07/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer stem cells (BCSCs) play essential roles in tumor formation, drug resistance, relapse, and metastasis. NANOG is a protein required for stem cell self-renewal, but the mechanisms by which it performs this function are poorly understood. Here, we show that hypoxia-inducible factor 1α (HIF-1α) is required for NANOG-mediated BCSC enrichment. Mechanistically, NANOG is recruited by HIF-1 to cooperatively activate transcription of the TERT gene encoding the telomerase reverse transcriptase that maintains telomere length, which is required for stem cell self-renewal. NANOG stimulates HIF-1 transcriptional activity by recruitment of the deubiquitinase USP9X, which inhibits HIF-1α protein degradation, and by stabilizing HIF-1α interaction with the coactivator p300, which mediates histone acetylation. Our results delineate a cooperative transcriptional mechanism by which HIF-1 and NANOG mediate BCSC self-renewal.
Collapse
|
22
|
Giunta S. Decoding human cancer with whole genome sequencing: a review of PCAWG Project studies published in February 2020. Cancer Metastasis Rev 2021; 40:909-924. [PMID: 34097189 PMCID: PMC8180541 DOI: 10.1007/s10555-021-09969-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022]
Abstract
Cancer is underlined by genetic changes. In an unprecedented international effort, the Pan-Cancer Analysis of Whole Genomes (PCAWG) of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA) sequenced the tumors of over two thousand five hundred patients across 38 different cancer types, as well as the corresponding healthy tissue, with the aim of identifying genome-wide mutations exclusively found in cancer and uncovering new genetic changes that drive tumor formation. What set this project apart from earlier efforts is the use of whole genome sequencing (WGS) that enabled to explore alterations beyond the coding DNA, into cancer's non-coding genome. WGS of the entire cohort allowed to tease apart driving mutations that initiate and support carcinogenesis from passenger mutations that do not play an overt role in the disease. At least one causative mutation was found in 95% of all cancers, with many tumors showing an average of 5 driver mutations. The PCAWG Project also assessed the transcriptional output altered in cancer and rebuilt the evolutionary history of each tumor showing that initial driver mutations can occur years if not decades prior to a diagnosis. Here, I provide a concise review of the Pan-Cancer Project papers published on February 2020, along with key computational tools and the digital framework generated as part of the project. This represents an historic effort by hundreds of international collaborators, which provides a comprehensive understanding of cancer genetics, with publicly available data and resources representing a treasure trove of information to advance cancer research for years to come.
Collapse
Affiliation(s)
- Simona Giunta
- Laboratory of Genome Evolution, Department of Biology & Biotechnology "Charles Darwin", University of Rome Sapienza, Rome, Italy.
- The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
23
|
Glenfield C, Innan H. Gene Duplication and Gene Fusion Are Important Drivers of Tumourigenesis during Cancer Evolution. Genes (Basel) 2021; 12:1376. [PMID: 34573358 PMCID: PMC8466788 DOI: 10.3390/genes12091376] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 02/07/2023] Open
Abstract
Chromosomal rearrangement and genome instability are common features of cancer cells in human. Consequently, gene duplication and gene fusion events are frequently observed in human malignancies and many of the products of these events are pathogenic, representing significant drivers of tumourigenesis and cancer evolution. In certain subsets of cancers duplicated and fused genes appear to be essential for initiation of tumour formation, and some even have the capability of transforming normal cells, highlighting the importance of understanding the events that result in their formation. The mechanisms that drive gene duplication and fusion are unregulated in cancer and they facilitate rapid evolution by selective forces akin to Darwinian survival of the fittest on a cellular level. In this review, we examine current knowledge of the landscape and prevalence of gene duplication and gene fusion in human cancers.
Collapse
Affiliation(s)
| | - Hideki Innan
- Department of Evolutionary Studies of Biosystems, SOKENDAI, The Graduate University for Advanced Studies, Shonan Village, Hayama, Kanagawar 240-0193, Japan;
| |
Collapse
|
24
|
da Silva EM, Selenica P, Vahdatinia M, Pareja F, Da Cruz Paula A, Ferrando L, Gazzo AM, Dopeso H, Ross DS, Bakhteri A, Riaz N, Chandarlapaty S, Razavi P, Norton L, Wen HY, Brogi E, Weigelt B, Zhang H, Reis-Filho JS. TERT promoter hotspot mutations and gene amplification in metaplastic breast cancer. NPJ Breast Cancer 2021; 7:43. [PMID: 33863915 PMCID: PMC8052452 DOI: 10.1038/s41523-021-00250-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Metaplastic breast cancers (MBCs) are characterized by complex genomes, which seem to vary according to their histologic subtype. TERT promoter hotspot mutations and gene amplification are rare in common forms of breast cancer, but present in a subset of phyllodes tumors. Here, we sought to determine the frequency of genetic alterations affecting TERT in a cohort of 60 MBCs with distinct predominant metaplastic components (squamous, 23%; spindle, 27%; osseous, 8%; chondroid, 42%), and to compare the repertoire of genetic alterations of MBCs according to the presence of TERT promoter hotspot mutations or gene amplification. Forty-four MBCs were subjected to: whole-exome sequencing (WES; n = 27) or targeted sequencing of 341-468 cancer-related genes (n = 17); 16 MBCs were subjected to Sanger sequencing of the TERT promoter, TP53 and selected exons of PIK3CA, HRAS, and BRAF. TERT promoter hotspot mutations (n = 9) and TERT gene amplification (n = 1) were found in 10 of the 60 MBCs analyzed, respectively. These TERT alterations were less frequently found in MBCs with predominant chondroid differentiation than in other MBC subtypes (p = 0.01, Fisher's exact test) and were mutually exclusive with TP53 mutations (p < 0.001, CoMEt). In addition, a comparative analysis of the MBCs subjected to WES or targeted cancer gene sequencing (n = 44) revealed that MBCs harboring TERT promoter hotspot mutations or gene amplification (n = 6) more frequently harbored PIK3CA than TERT wild-type MBCs (n = 38; p = 0.001; Fisher's exact test). In conclusion, TERT somatic genetic alterations are found in a subset of TP53 wild-type MBCs with squamous/spindle differentiation, highlighting the genetic diversity of these cancers.
Collapse
Affiliation(s)
- Edaise M da Silva
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mahsa Vahdatinia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnaud Da Cruz Paula
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lorenzo Ferrando
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Internal Medicine, University of Genoa, Genova, Italy
| | - Andrea M Gazzo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Higinio Dopeso
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ariya Bakhteri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
25
|
Dratwa M, Wysoczańska B, Łacina P, Kubik T, Bogunia-Kubik K. TERT-Regulation and Roles in Cancer Formation. Front Immunol 2020; 11:589929. [PMID: 33329574 PMCID: PMC7717964 DOI: 10.3389/fimmu.2020.589929] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is a catalytic subunit of telomerase. Telomerase complex plays a key role in cancer formation by telomere dependent or independent mechanisms. Telomere maintenance mechanisms include complex TERT changes such as gene amplifications, TERT structural variants, TERT promoter germline and somatic mutations, TERT epigenetic changes, and alternative lengthening of telomere. All of them are cancer specific at tissue histotype and at single cell level. TERT expression is regulated in tumors via multiple genetic and epigenetic alterations which affect telomerase activity. Telomerase activity via TERT expression has an impact on telomere length and can be a useful marker in diagnosis and prognosis of various cancers and a new therapy approach. In this review we want to highlight the main roles of TERT in different mechanisms of cancer development and regulation.
Collapse
Affiliation(s)
- Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Barbara Wysoczańska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Tomasz Kubik
- Department of Computer Engineering, Faculty of Electronics, Wrocław University of Science and Technology, Wroclaw, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
26
|
Prieto-Oliveira P. Telomerase activation in the treatment of aging or degenerative diseases: a systematic review. Mol Cell Biochem 2020; 476:599-607. [PMID: 33001374 DOI: 10.1007/s11010-020-03929-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Telomeres are protective structures that are shortened during the lifetime, resulting in aging and degenerative diseases. Subjects experiencing aging and degenerative disorders present smaller telomeres than young and healthy ones. The size of these structures can be stabilized by telomerase, an enzyme which is inactive in adult tissues but functional in fetal and newborn tissues and adult testes and ovaries. The aim of this study was to perform a systematic review to evaluate the effect of telomerase activation in the treatment of degenerative and aging disorders. We accomplished the search using the Pubmed interface for papers published from September 1985 to April 16th, 2020. We found twenty one studies that matched our eligibility criteria. I concluded that telomerase is probably a potential and safe treatment for aging and degenerative diseases, demonstrating neither side effects nor risk of cancer in the selected studies. Further studies in humans are needed to confirm safety and efficiency of this treatment.
Collapse
Affiliation(s)
- P Prieto-Oliveira
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Pedro de Toledo Street 781, 16th Floor, Retrovirology, Vila Clementino, São Paulo, SP, CEP: 04039-032, Brazil.
| |
Collapse
|
27
|
Faldoni FLC, Villacis RAR, Canto LM, Fonseca-Alves CE, Cury SS, Larsen SJ, Aagaard MM, Souza CP, Scapulatempo-Neto C, Osório CABT, Baumbach J, Marchi FA, Rogatto SR. Inflammatory Breast Cancer: Clinical Implications of Genomic Alterations and Mutational Profiling. Cancers (Basel) 2020; 12:cancers12102816. [PMID: 33007869 PMCID: PMC7650681 DOI: 10.3390/cancers12102816] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Inflammatory breast cancer (IBC) is an aggressive disease with high mortality rates. Nowadays, there is no targeted treatment for this tumor type. Based on this context, we investigated the molecular profile of this disease by using well-established methodologies (high-resolution microarray platform, targeted next-generation sequencing, and immunohistochemistry) that have proven potential to unveil cancer biomarkers. We found alterations related to IBC aggressiveness and metastasis (gains of MDM4, losses of CHL1, and high homologous recombination deficiency scores), and worse overall survival (variants in HR and mismatch repair genes). We also compared the mutational profiling of our cases with literature data, which includes both non-IBC and IBC cases, validating our findings. Overall, we describe genetic alterations with the potential to be used as prognostic or predictive biomarkers and ultimately improve IBC patients’ care. Abstract Inflammatory breast cancer (IBC) is a rare and aggressive type of breast cancer whose molecular basis is poorly understood. We performed a comprehensive molecular analysis of 24 IBC biopsies naïve of treatment, using a high-resolution microarray platform and targeted next-generation sequencing (105 cancer-related genes). The genes more frequently affected by gains were MYC (75%) and MDM4 (71%), while frequent losses encompassed TP53 (71%) and RB1 (58%). Increased MYC and MDM4 protein expression levels were detected in 18 cases. These genes have been related to IBC aggressiveness, and MDM4 is a potential therapeutic target in IBC. Functional enrichment analysis revealed genes associated with inflammatory regulation and immune response. High homologous recombination (HR) deficiency scores were detected in triple-negative and metastatic IBC cases. A high telomeric allelic imbalance score was found in patients having worse overall survival (OS). The mutational profiling was compared with non-IBC (TCGA, n = 250) and IBC (n = 118) from four datasets, validating our findings. Higher frequency of TP53 and BRCA2 variants were detected compared to non-IBC, while PIKC3A showed similar frequency. Variants in mismatch repair and HR genes were associated with worse OS. Our study provided a framework for improved diagnosis and therapeutic alternatives for this aggressive tumor type.
Collapse
Affiliation(s)
- Flávia L. C. Faldoni
- International Research Center, A.C.Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.L.C.F.); (F.A.M.)
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (L.M.C.); (M.M.A.)
| | - Rolando A. R. Villacis
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília-UnB, Brasília 70910-900, Brazil;
| | - Luisa M. Canto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (L.M.C.); (M.M.A.)
| | - Carlos E. Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu 18618-681, Brazil;
| | - Sarah S. Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University-UNESP, Botucatu 18618-689, Brazil;
| | - Simon J. Larsen
- Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark; (S.J.L.); (J.B.)
| | - Mads M. Aagaard
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (L.M.C.); (M.M.A.)
| | - Cristiano P. Souza
- Department of Breast and Gynecologic Oncology, Barretos Cancer Hospital, Pio XII Foundation, Barretos 14784-390, Brazil;
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos SP 14784-400, Brazil;
- Diagnósticos da América (DASA), Barueri 01525-001, Brazil
| | | | - Jan Baumbach
- Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark; (S.J.L.); (J.B.)
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Fabio A. Marchi
- International Research Center, A.C.Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.L.C.F.); (F.A.M.)
| | - Silvia R. Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (L.M.C.); (M.M.A.)
- Institute of Regional Health Research, University of Southern Denmark, 500 Odense, Denmark
- Correspondence:
| |
Collapse
|
28
|
TERT promotor region rearrangements analyzed in high-risk neuroblastomas by FISH method and whole genome sequencing. Int J Clin Oncol 2020; 25:2166-2174. [DOI: 10.1007/s10147-020-01773-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022]
|
29
|
Bradfield A, Button L, Drury J, Green DC, Hill CJ, Hapangama DK. Investigating the Role of Telomere and Telomerase Associated Genes and Proteins in Endometrial Cancer. Methods Protoc 2020; 3:E63. [PMID: 32899298 PMCID: PMC7565490 DOI: 10.3390/mps3030063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/24/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022] Open
Abstract
Endometrial cancer (EC) is the commonest gynaecological malignancy. Current prognostic markers are inadequate to accurately predict patient survival, necessitating novel prognostic markers, to improve treatment strategies. Telomerase has a unique role within the endometrium, whilst aberrant telomerase activity is a hallmark of many cancers. The aim of the current in silico study is to investigate the role of telomere and telomerase associated genes and proteins (TTAGPs) in EC to identify potential prognostic markers and therapeutic targets. Analysis of RNA-seq data from The Cancer Genome Atlas identified differentially expressed genes (DEGs) in EC (568 TTAGPs out of 3467) and ascertained DEGs associated with histological subtypes, higher grade endometrioid tumours and late stage EC. Functional analysis demonstrated that DEGs were predominantly involved in cell cycle regulation, while the survival analysis identified 69 DEGs associated with prognosis. The protein-protein interaction network constructed facilitated the identification of hub genes, enriched transcription factor binding sites and drugs that may target the network. Thus, our in silico methods distinguished many critical genes associated with telomere maintenance that were previously unknown to contribute to EC carcinogenesis and prognosis, including NOP56, WFS1, ANAPC4 and TUBB4A. Probing the prognostic and therapeutic utility of these novel TTAGP markers will form an exciting basis for future research.
Collapse
Affiliation(s)
- Alice Bradfield
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Lucy Button
- Faculty of Health and Life Sciences, University of Liverpool, Brownlow Hill, Liverpool L69 7ZX, UK;
| | - Josephine Drury
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Daniel C. Green
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, UK;
| | - Christopher J. Hill
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
| | - Dharani K. Hapangama
- Department of Women’s and Children’s Health, University of Liverpool, Crown St, Liverpool L69 7ZX, UK; (A.B.); (J.D.); (C.J.H.)
- Liverpool Women’s NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool L8 7SS, UK
| |
Collapse
|
30
|
Rossi T, Gallerani G, Angeli D, Cocchi C, Bandini E, Fici P, Gaudio M, Martinelli G, Rocca A, Maltoni R, Fabbri F. Single-Cell NGS-Based Analysis of Copy Number Alterations Reveals New Insights in Circulating Tumor Cells Persistence in Early-Stage Breast Cancer. Cancers (Basel) 2020; 12:cancers12092490. [PMID: 32887501 PMCID: PMC7565733 DOI: 10.3390/cancers12092490] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Circulating tumor cells (CTCs) are crucial for the identification of patients with a higher risk of relapse, including those diagnosed with breast cancer (BC). The aim of this study was to explore their molecular aspects in 11 early-stage BC patients during patient management, focusing on copy number alterations (CNAs) and exploiting a single-CTC next-generation sequencing approach. CTCs showed different degrees of aberration based on access time. Moreover, CTCs, in particular those persisting even months after tumor resection, shared CNAs with matched tumor tissue. Enrichment analyses of CNAs on CTCs highlighted peculiar aberrations, especially associated with interferon (IFN)-associated terms. The study of CTCs CNAs can provide information about the molecular mechanisms involving CTC-related processes and their survival ability in occult niches, supporting the goal of exploiting their application in patients’ surveillance and follow-up. Abstract Circulating tumor cells (CTCs) are a rare population of cells representing a key player in the metastatic cascade. They are recognized as a validated tool for the identification of patients with a higher risk of relapse, including those diagnosed with breast cancer (BC). However, CTCs are characterized by high levels of heterogeneity that also involve copy number alterations (CNAs), structural variations associated with gene dosage changes. In this study, single CTCs were isolated from the peripheral blood of 11 early-stage BC patients at different time points. A label-free enrichment of CTCs was performed using OncoQuick, and single CTCs were isolated using DEPArray. Libraries were prepared from single CTCs and DNA extracted from matched tumor tissues for a whole-genome low-coverage next-generation sequencing (NGS) analysis using the Ion Torrent S5 System. The analysis of the CNA burden highlighted that CTCs had different degrees of aberration based on the time point and subtype. CTCs were found even six months after surgery and shared CNAs with matched tumor tissue. Tumor-associated CNAs that were recurrent in CTCs were patient-specific, and some alterations involved regions associated with BC and survival (i.e., gains at 1q21-23 and 5p15.33). The enrichment analysis emphasized the involvement of aberrations of terms, associated in particular with interferon (IFN) signaling. Collectively, our findings reveal that these aberrations may contribute to understanding the molecular mechanisms involving CTC-related processes and their survival ability in occult niches, supporting the goal of exploiting their application in patients’ surveillance and follow-up.
Collapse
Affiliation(s)
- Tania Rossi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
- Correspondence: ; Tel.: +39-05-4373-9982
| | - Giulia Gallerani
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Claudia Cocchi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
| | - Erika Bandini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
| | - Pietro Fici
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
| | - Michele Gaudio
- Pathology Unit, AUSL Romagna, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy;
| | - Giovanni Martinelli
- Scientific Directorate, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Andrea Rocca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.R.); (R.M.)
| | - Roberta Maltoni
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.R.); (R.M.)
| | - Francesco Fabbri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.G.); (C.C.); (E.B.); (P.F.); (F.F.)
| |
Collapse
|
31
|
Kim TM, Yang IS, Seung BJ, Lee S, Kim D, Ha YJ, Seo MK, Kim KK, Kim HS, Cheong JH, Sur JH, Nam H, Kim S. Cross-species oncogenic signatures of breast cancer in canine mammary tumors. Nat Commun 2020; 11:3616. [PMID: 32680987 PMCID: PMC7367841 DOI: 10.1038/s41467-020-17458-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/02/2020] [Indexed: 02/08/2023] Open
Abstract
Genomic and precision medicine research has afforded notable advances in human cancer treatment, yet applicability to other species remains uncertain. Through whole-exome and transcriptome analyses of 191 spontaneous canine mammary tumors (CMTs) that exhibit the archetypal features of human breast cancers, we found a striking resemblance of genomic characteristics including frequent PIK3CA mutations (43.1%), aberrations of the PI3K-Akt pathway (61.7%), and key genes involved in cancer initiation and progression. We also identified three gene expression-based CMT subtypes, one of which segregated with basal-like human breast cancer subtypes with activated epithelial-to-mesenchymal transition, low claudin expression, and unfavorable disease prognosis. A relative lack of ERBB2 amplification and Her2-enrichment subtype in CMT denoted species-specific molecular mechanisms. Taken together, our results elucidate cross-species oncogenic signatures for a better understanding of universal and context-dependent mechanisms in breast cancer development and provide a basis for precision diagnostics and therapeutics for domestic dogs.
Collapse
Affiliation(s)
- Tae-Min Kim
- Department of Medical Informatics and Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - In Seok Yang
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Byung-Joon Seung
- Department of Veterinary Pathology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Sejoon Lee
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Dohyun Kim
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Yoo-Jin Ha
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Mi-Kyoung Seo
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Ka-Kyung Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyun Seok Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jae-Ho Cheong
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jung-Hyang Sur
- Department of Veterinary Pathology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, South Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
32
|
Srinivas N, Rachakonda S, Kumar R. Telomeres and Telomere Length: A General Overview. Cancers (Basel) 2020; 12:E558. [PMID: 32121056 PMCID: PMC7139734 DOI: 10.3390/cancers12030558] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
Telomeres are highly conserved tandem nucleotide repeats that include proximal double-stranded and distal single-stranded regions that in complex with shelterin proteins afford protection at chromosomal ends to maintain genomic integrity. Due to the inherent limitations of DNA replication and telomerase suppression in most somatic cells, telomeres undergo age-dependent incremental attrition. Short or dysfunctional telomeres are recognized as DNA double-stranded breaks, triggering cells to undergo replicative senescence. Telomere shortening, therefore, acts as a counting mechanism that drives replicative senescence by limiting the mitotic potential of cells. Telomere length, a complex hereditary trait, is associated with aging and age-related diseases. Epidemiological data, in general, support an association with varying magnitudes between constitutive telomere length and several disorders, including cancers. Telomere attrition is also influenced by oxidative damage and replicative stress caused by genetic, epigenetic, and environmental factors. Several single nucleotide polymorphisms at different loci, identified through genome-wide association studies, influence inter-individual variation in telomere length. In addition to genetic factors, environmental factors also influence telomere length during growth and development. Telomeres hold potential as biomarkers that reflect the genetic predisposition together with the impact of environmental conditions and as targets for anti-cancer therapies.
Collapse
Affiliation(s)
| | | | - Rajiv Kumar
- Division of Functional Genome Analysis, German Cancer Research Center, Im Neunheimer Feld 580, 69120 Heidelberg, Germany; (N.S.); (S.R.)
| |
Collapse
|
33
|
Lopes TZ, de Moraes FR, Tedesco AC, Arni RK, Rahal P, Calmon MF. Berberine associated photodynamic therapy promotes autophagy and apoptosis via ROS generation in renal carcinoma cells. Biomed Pharmacother 2019; 123:109794. [PMID: 31874443 DOI: 10.1016/j.biopha.2019.109794] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) consists of the most lethal common urological cancer and the clinical practice has shown that resistant RCC to commons therapies is extremely high. Berberine is an isoquinoline alkaloid, presents in different kinds of plants and it has long been used in Chinese medicine. It has several properties, such as antioxidant, anti-inflammatory, anti-diabetic, anti-microbial and anti-cancer. Moreover, berberine has photosensitive characteristics and its association with photodynamic therapy (PDT) is effective against tumor cells. This study aimed to evaluate the effects of berberine associated with PDT in renal carcinoma cell lines. The cellular viability assay showed increased cytotoxicity in concentration and time-dependent manner. Berberine presented efficient internalization in all cell lines analyzed. In addition, after treatment with berberine associated with PDT, it was observed a high phototoxicity effect with less than 20 % of viable cells. In this study we observed that the increase of reactive oxygen species (ROS) levels was accompanied by an increase of autophagy levels and apoptosis by caspase 3 activity, suggesting cell death by both mechanisms. Additionally, three target genes of anti-cancer drugs were differentially expressed in 786-O cells, being that Vascular Endothelial Growth Factor-D (FIGF) and Human Telomerase Reverse Transcriptase (TERT) gene presented low expression and Polo Like Kinase 3 (PLK3) presented overexpression after treatment with berberine associated with PDT. In this study, the proposed treatment triggered metabolites changes related to cell proliferation, tumorigenesis and angiogenesis. Thus, it was possible to suggest that berberine has promising potential as a photosensitizing agent in a photodynamic therapy, because it induced significant anticancer effects on renal carcinoma cells.
Collapse
Affiliation(s)
- Tairine Zara Lopes
- Laboratory of Genomics Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Fabio Rogério de Moraes
- Physics Department, São Paulo State University, São José do Rio Preto, São Paulo, Brazil; Multiuser Center for Biomolecular Innovation, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Raghuvir Krishnaswamy Arni
- Physics Department, São Paulo State University, São José do Rio Preto, São Paulo, Brazil; Multiuser Center for Biomolecular Innovation, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Paula Rahal
- Laboratory of Genomics Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Marilia Freitas Calmon
- Laboratory of Genomics Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
34
|
Rahman MM, Brane AC, Tollefsbol TO. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer. Cells 2019; 8:cells8101214. [PMID: 31597272 PMCID: PMC6829616 DOI: 10.3390/cells8101214] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/04/2019] [Accepted: 10/06/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is a sporadic disease with genetic and epigenetic components. Genomic instability in breast cancer leads to mutations, copy number variations, and genetic rearrangements, while epigenetic remodeling involves alteration by DNA methylation, histone modification and microRNAs (miRNAs) of gene expression profiles. The accrued scientific findings strongly suggest epigenetic dysregulation in breast cancer pathogenesis though genomic instability is central to breast cancer hallmarks. Being reversible and plastic, epigenetic processes appear more amenable toward therapeutic intervention than the more unidirectional genetic alterations. In this review, we discuss the epigenetic reprogramming associated with breast cancer such as shuffling of DNA methylation, histone acetylation, histone methylation, and miRNAs expression profiles. As part of this, we illustrate how epigenetic instability orchestrates the attainment of cancer hallmarks which stimulate the neoplastic transformation-tumorigenesis-malignancy cascades. As reversibility of epigenetic controls is a promising feature to optimize for devising novel therapeutic approaches, we also focus on the strategies for restoring the epistate that favor improved disease outcome and therapeutic intervention.
Collapse
Affiliation(s)
- Mohammad Mijanur Rahman
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Andrew C Brane
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Center for Healthy Aging, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA.
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA.
| |
Collapse
|
35
|
Panebianco F, Nikitski AV, Nikiforova MN, Nikiforov YE. Spectrum of TERT promoter mutations and mechanisms of activation in thyroid cancer. Cancer Med 2019; 8:5831-5839. [PMID: 31408918 PMCID: PMC6792496 DOI: 10.1002/cam4.2467] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Reactivation of telomerase reverse transcriptase (TERT) is an important event in cancer. Two hotspot mutations in the TERT promoter region, c.-124C > T (C228T) and c.-146C > T (C250T), occur in various cancer types including thyroid cancer. They generate de novo binding sites for E-twenty-six (ETS) transcription factors causing increased TERT transcription. The aim of this study was to search for novel TERT promoter mutations and additional mechanisms of TERT activation in thyroid cancer. METHODS We studied 198 papillary thyroid carcinomas (PTCs), 34 follicular thyroid carcinomas (FTCs), 40 Hürthle cell carcinomas (HCCs), 14 poorly differentiated/anaplastic thyroid carcinomas (PDTC/ATC), and 15 medullary thyroid carcinomas (MTCs) for mutations in an -424 bp to +64 bp region of TERT. The luciferase reporter assay was used to functionally characterize the identified alterations. Copy number variations (CNVs) in the TERT region were analyzed using TaqMan copy number assay and validated with fluorescence in situ hybridization (FISH). RESULTS We detected the hotspot c.-124C > T and c.-146C > T mutations in 7% PTC, 18% FTC, 25% HCC, and 86% PDTC/ATC. One PTC carried a c.-124C > A mutation. Furthermore, we identified two novel mutations resulting in the formation of de novo ETS-binding motifs: c.-332C > T in one MTC and c.-104_-83dup in one PTC. These genetic alterations, as well as other detected mutations, led to a significant increase in TERT promoter activity when assayed using luciferase reporter system. In addition, 5% of thyroid tumors were found to have ≥3 copies of TERT. CONCLUSIONS This study confirms the increased prevalence of TERT promoter mutations and CNV in advanced thyroid cancers and describes novel functional alterations in the TERT gene promoter, including a point mutation and small duplication. These mutations, as well as TERT copy number alterations, may represent an additional mechanism of TERT activation in thyroid cancer.
Collapse
Affiliation(s)
- Federica Panebianco
- Department of Pathology and Laboratory MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Alyaksandr V. Nikitski
- Department of Pathology and Laboratory MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Marina N. Nikiforova
- Department of Pathology and Laboratory MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| | - Yuri E. Nikiforov
- Department of Pathology and Laboratory MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvania
| |
Collapse
|
36
|
Rowland TJ, Dumbović G, Hass EP, Rinn JL, Cech TR. Single-cell imaging reveals unexpected heterogeneity of telomerase reverse transcriptase expression across human cancer cell lines. Proc Natl Acad Sci U S A 2019; 116:18488-18497. [PMID: 31451652 PMCID: PMC6744858 DOI: 10.1073/pnas.1908275116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Telomerase is pathologically reactivated in most human cancers, where it maintains chromosomal telomeres and allows immortalization. Because telomerase reverse transcriptase (TERT) is usually the limiting component for telomerase activation, numerous studies have measured TERT mRNA levels in populations of cells or in tissues. In comparison, little is known about TERT expression at the single-cell and single-molecule level. To address this, we analyzed TERT expression across 10 human cancer lines using single-molecule RNA fluorescent in situ hybridization (FISH) and made several unexpected findings. First, there was substantial cell-to-cell variation in number of transcription sites and ratio of transcription sites to gene copies. Second, previous classification of lines as having monoallelic or biallelic TERT expression was found to be inadequate for capturing the TERT gene expression patterns. Finally, spliced TERT mRNA had primarily nuclear localization in cancer cells and induced pluripotent stem cells (iPSCs), in stark contrast to the expectation that spliced mRNA should be predominantly cytoplasmic. These data reveal unappreciated heterogeneity, complexity, and unconventionality in TERT expression across human cancer cells.
Collapse
Affiliation(s)
- Teisha J Rowland
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Gabrijela Dumbović
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Evan P Hass
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - John L Rinn
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Thomas R Cech
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303;
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO 80303
| |
Collapse
|
37
|
Lee DD, Leão R, Komosa M, Gallo M, Zhang CH, Lipman T, Remke M, Heidari A, Nunes NM, Apolónio JD, Price AJ, De Mello RA, Dias JS, Huntsman D, Hermanns T, Wild PJ, Vanner R, Zadeh G, Karamchandani J, Das S, Taylor MD, Hawkins CE, Wasserman JD, Figueiredo A, Hamilton RJ, Minden MD, Wani K, Diplas B, Yan H, Aldape K, Akbari MR, Danesh A, Pugh TJ, Dirks PB, Castelo-Branco P, Tabori U. DNA hypermethylation within TERT promoter upregulates TERT expression in cancer. J Clin Invest 2018; 129:223-229. [PMID: 30358567 DOI: 10.1172/jci121303] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023] Open
Abstract
Replicative immortality is a hallmark of cancer cells governed by telomere maintenance. Approximately 90% of human cancers maintain their telomeres by activating telomerase, driven by the transcriptional upregulation of telomerase reverse transcriptase (TERT). Although TERT promoter mutations (TPMs) are a major cancer-associated genetic mechanism of TERT upregulation, many cancers exhibit TERT upregulation without TPMs. In this study, we describe the TERT hypermethylated oncological region (THOR), a 433-bp genomic region encompassing 52 CpG sites located immediately upstream of the TERT core promoter, as a cancer-associated epigenetic mechanism of TERT upregulation. Unmethylated THOR repressed TERT promoter activity regardless of TPM status, and hypermethylation of THOR counteracted this repressive function. THOR methylation analysis in 1,352 human tumors revealed frequent (>45%) cancer-associated DNA hypermethylation in 9 of 11 (82%) tumor types screened. Additionally, THOR hypermethylation, either independently or along with TPMs, accounted for how approximately 90% of human cancers can aberrantly activate telomerase. Thus, we propose that THOR hypermethylation is a prevalent telomerase-activating mechanism in cancer that can act independently of or in conjunction with TPMs, further supporting the utility of THOR hypermethylation as a prognostic biomarker.
Collapse
Affiliation(s)
- Donghyun D Lee
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ricardo Leão
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Martin Komosa
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marco Gallo
- Departments of Physiology and Pharmacology, Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Cindy H Zhang
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tatiana Lipman
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marc Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Abolfazl Heidari
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nuno Miguel Nunes
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joana D Apolónio
- Department of Biomedical Sciences and Medicine, and.,Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - Aryeh J Price
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - João S Dias
- Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - David Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Peter J Wild
- Institute of Pathology and Molecular Pathology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Robert Vanner
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Division of Neurosurgery, University of Toronto, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jason Karamchandani
- Department of Pathology, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Sunit Das
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cynthia E Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jonathan D Wasserman
- Division of Endocrinology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Robert J Hamilton
- Division of Urology, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Khalida Wani
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bill Diplas
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Hai Yan
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Kenneth Aldape
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mohammad R Akbari
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Arnavaz Danesh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Peter B Dirks
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Uri Tabori
- Program in Genetics and Genome Biology, and.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Genomic profiling of metaplastic breast carcinomas reveals genetic heterogeneity and relationship to ductal carcinoma. Mod Pathol 2018; 31:1661-1674. [PMID: 29946183 DOI: 10.1038/s41379-018-0081-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/30/2022]
Abstract
Metaplastic breast carcinomas comprise a histologically heterogenous group of tumors. Although most are triple (estrogen/progesterone receptor, HER2) negative, these rare tumors are clinicopathologically distinct from other triple negative carcinomas and may be aggressive with worse chemotherapy responses. On the other hand, metaplastic carcinomas are histologically diverse, which is reflected in gene expression differences among subtypes. Whether metaplastic carcinomas are genetically distinct from other triple negative cancers and whether genetic differences underlie histologic subtypes remains poorly understood. We sequenced 408 cancer-related genes in 28 metaplastic carcinomas, including chondroid matrix-producing carcinomas (n = 10), spindle cell carcinomas (n = 5), and carcinomas with squamous (n = 5), mixed spindle/squamous (n = 5), and mixed metaplastic (n = 3) differentiation. Metaplastic carcinomas were highly enriched for PIK3CA/PIK3R1 (61%) and Ras-Map kinase (25%) pathway aberrations compared to other triple negative carcinomas (TCGA dataset 14%, p < 0.001 and 7%, p = 0.005, respectively) and harbored a high frequency of TP53 (64%) and TERT promoter (25%) mutations, but this varied among subtypes. Chondroid-matrix producing carcinomas lacked PI-3 kinase and Ras-Map kinase aberrations and TERT promoter mutations, compared to 100%, 39%, and 39% of non-matrix-producing tumors, respectively. TERT promoter mutations were enriched (47%) in spindle cell carcinomas and tumors with squamous or spindle/squamous differentiation. Spindle cell carcinomas lacked TP53 mutations, in contrast to other subtypes (78%, p = 0.003). Separate analysis of paired ductal carcinoma in situ and metaplastic carcinoma revealed shared clonality in all cases (n = 8). Activating PI-3 kinase and Ras pathway mutations were early events, and inactivating mutations in tumor suppressors including RB1, CDKN2A, and TP53 were associated with invasion in individual cases. Metaplastic components of two tumors showed genetic progression from separately sequenced paired invasive ductal carcinoma. The findings suggest that metaplastic carcinomas are genetically distinct from other triple negative breast cancers and highlight genetic heterogeneity that broadly correlates with histologic subtype. Heterologous elements progress from associated ductal carcinoma.
Collapse
|
39
|
Vizcaino MA, Palsgrove DN, Yuan M, Giannini C, Cabrera-Aldana EE, Pallavajjala A, Burger PC, Rodriguez FJ. Granular cell astrocytoma: an aggressive IDH-wildtype diffuse glioma with molecular genetic features of primary glioblastoma. Brain Pathol 2018; 29:193-204. [PMID: 30222900 DOI: 10.1111/bpa.12657] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022] Open
Abstract
Granular cell astrocytoma (GCA) is a rare adult infiltrating glioma subtype. We studied a series of 39 GCAs. Median age of presentation was 57.8 years and most cases developed in the frontal or temporal lobes. Tumors included grade II (n = 14), grade III (n = 11), and grade IV (n = 14) by WHO criteria. Granular cell morphology was diffuse in 31 (79%) cases and partial in eight (21%). Immunohistochemistry showed frequent positivity for GFAP (28 of 31), OLIG2 (16 of 16), and CD68 (27 of 30), but HAM56, CD163, and IBA-1 histiocytic markers were all negative (22 of 22). IDH1(R132H) was negative in all the cases tested (16 of 16), while ATRX expression was retained (12 of 12). Cytogenetics demonstrated monosomy 10 (6 of 6) cases, +7 in 4 (of 6), -13q in 4 of 6, and -14 in 4 of 6. Next-generation sequencing demonstrated mutations in PTEN/PIK3 genes in 6/13 (46%), NF1 in 3 of 10 (30%), TP53 in 3 of 13 (23%), PALB2 in 3 of 10 (30%), STAG2 in 3 of 10 (30%), EGFR mutation/amplification in 3 of 13 (23%), and AR in 2 of 10 (20%). CDKN2A/B deletion was identified in 5 of 13 (30%) cases (homozygous deletion in 4). The TERT C228T mutation was identified in 9 of 13 (69%). No mutations were encountered in IDH1, IDH2, CIC, FUBP1, H3F3A, BRAF or ATRX genes. The mean overall survival was 11.3 months. Patients >60 years old at diagnosis had a worse survival than patients <60 years (P = 0.001). There were no statistically significant differences in survival by WHO grade, extent of granular cell change, sex or MIB-1 (P > 0.05). GCA is a variant of IDH-wildtype diffuse glioma with aggressive behavior irrespective of grade and extent of granular cell morphology, and with molecular genetic features corresponding to primary glioblastoma.
Collapse
Affiliation(s)
- M Adelita Vizcaino
- Department of Pathology, Johns Hopkins University, Baltimore, MD.,Faculty of Medicine, Department of Cellular and Tissue Biology, UNAM, Mexico City, Mexico
| | - Doreen N Palsgrove
- Department of Pathology, Johns Hopkins University, Baltimore, MD.,Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Ming Yuan
- Department of Pathology, Johns Hopkins University, Baltimore, MD
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | | | - Peter C Burger
- Department of Pathology, Johns Hopkins University, Baltimore, MD.,Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University, Baltimore, MD.,Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
40
|
Gaspar TB, Sá A, Lopes JM, Sobrinho-Simões M, Soares P, Vinagre J. Telomere Maintenance Mechanisms in Cancer. Genes (Basel) 2018; 9:E241. [PMID: 29751586 PMCID: PMC5977181 DOI: 10.3390/genes9050241] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/12/2022] Open
Abstract
Tumour cells can adopt telomere maintenance mechanisms (TMMs) to avoid telomere shortening, an inevitable process due to successive cell divisions. In most tumour cells, telomere length (TL) is maintained by reactivation of telomerase, while a small part acquires immortality through the telomerase-independent alternative lengthening of telomeres (ALT) mechanism. In the last years, a great amount of data was generated, and different TMMs were reported and explained in detail, benefiting from genome-scale studies of major importance. In this review, we address seven different TMMs in tumour cells: mutations of the TERT promoter (TERTp), amplification of the genes TERT and TERC, polymorphic variants of the TERT gene and of its promoter, rearrangements of the TERT gene, epigenetic changes, ALT, and non-defined TMM (NDTMM). We gathered information from over fifty thousand patients reported in 288 papers in the last years. This wide data collection enabled us to portray, by organ/system and histotypes, the prevalence of TERTp mutations, TERT and TERC amplifications, and ALT in human tumours. Based on this information, we discuss the putative future clinical impact of the aforementioned mechanisms on the malignant transformation process in different setups, and provide insights for screening, prognosis, and patient management stratification.
Collapse
Affiliation(s)
- Tiago Bordeira Gaspar
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Medical Faculty of University of Porto (FMUP), 4200-139 Porto, Portugal.
- Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| | - Ana Sá
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| | - José Manuel Lopes
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Medical Faculty of University of Porto (FMUP), 4200-139 Porto, Portugal.
- Department of Pathology and Oncology, Centro Hospitalar São João, 4200-139 Porto, Portugal.
| | - Manuel Sobrinho-Simões
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Medical Faculty of University of Porto (FMUP), 4200-139 Porto, Portugal.
- Department of Pathology and Oncology, Centro Hospitalar São João, 4200-139 Porto, Portugal.
| | - Paula Soares
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| | - João Vinagre
- Cancer Signaling and Metabolism Group, Institute for Research and Innovation in Health Sciences (i3S), University of Porto, 4200-135 Porto, Portugal.
- Cancer Signaling and Metabolism Group, Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), 4200-135 Porto, Portugal.
- Medical Faculty of University of Porto (FMUP), 4200-139 Porto, Portugal.
| |
Collapse
|