1
|
Herreros‐Pomares A, Hervás D, Bagán L, Proaño A, Bagan J. Proliferative verrucous and homogeneous Leukoplakias exhibit differential methylation patterns. Oral Dis 2025; 31:137-147. [PMID: 38852153 PMCID: PMC11808169 DOI: 10.1111/odi.15028] [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: 11/13/2023] [Revised: 03/20/2024] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVE Proliferative verrucous leukoplakia (PVL) is considered a clinically distinct entity from other oral leucoplakias (OLs) due to its clinical presentation and evolution. However, molecular differences between them remain unclear. We aimed to determine whether there are methylation differences between PVL and other forms of OLs. MATERIALS AND METHODS Oral biopsies from 12 patients with PVL, eight patients with homogeneous leucoplakia (HL), and 10 healthy individuals were obtained for a genome-wide DNA methylation analysis via the Infinium EPIC Platform. RESULTS A total of 1815 differentially methylated CpGs were found between PVL and HL, with a prominent state of hypermethylation in HL patients. CpGs covered 813 genes with distinct roles, including cell adhesion, extracellular matrix organization, and cell and synaptic signaling. 43% of these genes had been previously described in cancer and associated with prognosis. We developed a multinomial logistic regression model able to differentiate HL, PVL, and control samples. The model had a cross-validated estimate of 73% and included differentially methylated cancer-related genes between the pathological conditions and the healthy donors, including ADNP, BRCA2, CDK13, GNB1, NIN, NUMB, PIK3C2B, PTK2, SHISA4, THSD7B, WWP1, and ZNF292. It also included CpGs covering differentially methylated genes in HL (MEN1 and TNRC6B) and PVL (ACOXL, ADH1B, CAMTA1, CBFA2T3, CPXM2, LRFN2, SORCS2, and SPN). CONCLUSIONS PVL and HL present differential methylation patterns that could be linked to their differential clinical behavior. Our findings show the potential of methylation markers and suggest novel diagnostic biomarkers.
Collapse
Affiliation(s)
- Alejandro Herreros‐Pomares
- Department of BiotechnologyUniversitat Politècnica de ValènciaValenciaSpain
- Centro de Investigación Biomédica en Red CáncerCIBERONCMadridSpain
| | - David Hervás
- Departament of Applied Statistics and Operational Research, and QualityUniversitat Politècnica de ValènciaValenciaSpain
| | - Leticia Bagán
- Medicina Oral Unit, Stomatology DepartmentValencia UniversityValenciaSpain
| | - Alex Proaño
- Medicina Oral Unit, Stomatology DepartmentValencia UniversityValenciaSpain
| | - José Bagan
- Centro de Investigación Biomédica en Red CáncerCIBERONCMadridSpain
- Medicina Oral Unit, Stomatology DepartmentValencia UniversityValenciaSpain
- Department of Stomatology and Maxillofacial SurgeryHospital General Universitario de ValenciaValenciaSpain
- Precancer and Oral Cancer Research Group of Valencia UniversityValenciaSpain
| |
Collapse
|
2
|
Gabusi A, Gissi DB, Querzoli G, Sangiovanni A, Rossi R, Lucchi E, Tarsitano A, Montebugnoli L, Foschini MP, Morandi L. DNA methylation analysis from oral brushing reveals a field cancerization effect in proliferative verrucous leukoplakia. Pathologica 2024; 116:368-378. [PMID: 39748722 DOI: 10.32074/1591-951x-n838] [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: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Objectives The aim of the present study was to analyze the methylation status in patients who presented with an Oral Squamous Cell Carcinoma (OSCC) concomitantly with multifocal Proliferative Verrucous Leukoplakia (PVL)(PVL-OSCC). Methods Nine patients with OSCC and concomitant PVL lesions were selected. Two brushing samples were collected simultaneously from OSCC and PVL lesions in contralateral mucosa from each patient. 15 genes (272 CpGs) were used to compare methylation profiles of PVL-OSCC and paired OSCC. CpGs with a methylation level superimposable between PVL-OSCC and contralateral OSCC were selected for a comparative analysis between PVL-OSCC, 8 PVL patients with no history of OSCC (PVL) and 23 healthy donors. Samples were also tested using an algorithm that was recently validated for epigenetic alterations in OSCC. Results 220/272 CpGs islands (80%) showed a superimposable methylation level in OSCC and in PVL-OSCC. 10 genes (88 CpGs) and in particular PARP15 and ITGA4 (100% of the studied CpGs) were able to stratify PVL-OSCC from PVL and healthy donors. 3/4 (75%) PVL-OSCC patients with a "positive" algorithm score developed second neoplastic events compared to only 1/5 (20%) patients with a "negative" score. Conclusions The present study provides evidence that PVL shares an aberrant methylation profile with contralateral OSCC. In agreement with the theory of field cancerization, our data point towards the potential role of epigenetics in patients at risk of developing multiple neoplastic events.
Collapse
Affiliation(s)
- Andrea Gabusi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Davide Bartolomeo Gissi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Giulia Querzoli
- Section of Anatomic Pathology S. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Asia Sangiovanni
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Roberto Rossi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Lucchi
- Oral and Maxillofacial Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Achille Tarsitano
- Oral and Maxillofacial Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, Section of Maxillo-facial Surgery at Policlinico S. Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Lucio Montebugnoli
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Maria Pia Foschini
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology at Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Luca Morandi
- Functional and Molecular Neuroimaging Unit, Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
3
|
Pellegrini C, Ravaioli F, De Fanti S, Pirazzini C, D’Silva C, Garagnani P, Franceschi C, Bonifazi F, Zinzani PL, Bonafè M, Guarino M, Lodi R, Cortelli P, Tonon C, Mitolo M, Sambati L, Morandi L, Bacalini MG. Detection of Brain-Derived Cell-Free DNA in Plasma. Diagnostics (Basel) 2024; 14:2541. [PMID: 39594207 PMCID: PMC11592591 DOI: 10.3390/diagnostics14222541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Neuronal loss is a major pathological feature of neurodegenerative diseases. The analysis of plasma cell-free DNA (cfDNA) is an emerging approach to track cell death events in a minimally invasive way and from inaccessible areas of the body, such as the brain. Previous studies showed that DNA methylation (DNAm) profiles can be used to map the tissue of origin of cfDNA and to identify molecules released from the brain upon cell death. The aim of the present study is to contribute to this research field, presenting the development and validation of an assay for the detection of brain-derived cfDNA (bcfDNA). Methods: To identify CpG sites with brain-specific DNAm, we compared brain and non-brain tissues for their chromatin state profiles and genome-wide DNAm data, available in public datasets. The selected target genomic regions were experimentally validated by bisulfite sequencing on DNA extracted from 44 different autoptic tissues, including multiple brain regions. Sequencing data were analysed to identify brain-specific epihaplotypes. The developed assay was tested in plasma cfDNA from patients with immune effector cell-associated neurotoxicity syndrome (ICANS) following chimeric antigen receptor T (CAR-T) therapy. Results: We validated five genomic regions with brain-specific DNAm (four hypomethylated and one hypermethylated in the brain). DNAm analysis of the selected genomic regions in plasma samples from CAR-T patients revealed higher levels of bcfDNA in participants with ongoing neurotoxicity syndrome. Conclusions: We developed an assay for the analysis of bcfDNA in plasma. The assay is a promising tool for the early detection of neuronal loss in neurodegenerative diseases.
Collapse
Affiliation(s)
- Camilla Pellegrini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| | - Francesco Ravaioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| | - Sara De Fanti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| | - Chiara Pirazzini
- Department of Medical and Surgical Sciences—DIMEC, University of Bologna, 40126 Bologna, Italy; (C.P.); (P.G.); (P.L.Z.); (M.B.)
| | - Chiara D’Silva
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy;
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences—DIMEC, University of Bologna, 40126 Bologna, Italy; (C.P.); (P.G.); (P.L.Z.); (M.B.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging, Institute of Biology and Biomedicine and Institute of Information Technology, Mathematics and Mechanics, Department of Applied Mathematics, Lobachevsky State University, 603950 Nizhny Novgorod, Russia;
| | - Francesca Bonifazi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Pier Luigi Zinzani
- Department of Medical and Surgical Sciences—DIMEC, University of Bologna, 40126 Bologna, Italy; (C.P.); (P.G.); (P.L.Z.); (M.B.)
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Massimiliano Bonafè
- Department of Medical and Surgical Sciences—DIMEC, University of Bologna, 40126 Bologna, Italy; (C.P.); (P.G.); (P.L.Z.); (M.B.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| | - Raffaele Lodi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Pietro Cortelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Caterina Tonon
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Micaela Mitolo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Luisa Sambati
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| | - Luca Morandi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy
| | - Maria Giulia Bacalini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (C.P.); (F.R.); (S.D.F.); (M.G.); (R.L.); (P.C.); (C.T.); (L.S.); (L.M.)
| |
Collapse
|
4
|
Rapado-González Ó, Salta S, López-López R, Henrique R, Suárez-Cunqueiro MM, Jerónimo C. DNA methylation markers for oral cancer detection in non- and minimally invasive samples: a systematic review. Clin Epigenetics 2024; 16:105. [PMID: 39138540 PMCID: PMC11323632 DOI: 10.1186/s13148-024-01716-9] [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: 04/26/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
More than 50% of oral cancer (OC) patients are diagnosed with advanced-stage disease associated with poor prognosis and quality of life, supporting an urgent need to improve early OC detection. The identification of effective molecular markers by minimally invasive approaches has emerged as a promising strategy for OC screening. This systematic review summarizes and evaluates the performance of the DNA methylation markers identified in non- or minimally invasive samples for OC detection. PubMed's MEDLINE, Scopus, Embase, and Cochrane Library databases were systematically searched for studies that evaluated DNA methylation markers in non-invasive and/or minimally invasive samples (oral rinse/saliva, oral brush, and blood) from OC patients. Two investigators independently extracted data on study population characteristics, candidate methylation markers, testing samples, DNA methylation assay, and performance diagnostic outcomes. Methodological study quality was assessed with the Quality Assessment for Studies of Diagnostic Accuracy-2 tool. Thirty-one studies met the inclusion criteria for this systematic review. DNA methylation markers were evaluated in oral rinse/saliva (n = 17), oral brush (n = 9), and blood (n = 7) samples. Methylation-specific PCR (MSP) and quantitative-MSP were the most common DNA methylation assays. Regarding diagnostic performance values for salivary, oral brush, and blood DNA methylation markers, sensitivity and specificity ranged between 3.4-100% and 21-100%, 9-100% and 26.8-100%, 22-70% and 45.45-100%, respectively. Different gene methylation panels showed good diagnostic performance for OC detection. This systematic review discloses the promising value of testing DNA methylation markers in non-invasive (saliva or oral rinse) or minimally invasive (oral brush or blood) samples as a novel strategy for OC detection. However, further validation in large, multicenter, and prospective study cohorts must be carried out to confirm the clinical value of specific DNA methylation markers in this setting.
Collapse
Affiliation(s)
- Óscar Rapado-González
- Department of Surgery and Medical-Surgical Specialties, Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), 15706, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Sofia Salta
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Rafael López-López
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS, SERGAS), 15706, Santiago de Compostela, Spain
| | - Rui Henrique
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| | - María Mercedes Suárez-Cunqueiro
- Department of Surgery and Medical-Surgical Specialties, Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School, Universidade de Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS, SERGAS), 15706, Santiago de Compostela, Spain
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center - Raquel Seruca (Porto.CCC) & CI-IPOP@RISE (Health Research Network), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
5
|
Yang X, Du G, Ji T, Shi L, Liu W. Focus on DNA methylation in saliva and oral swabs for oral potentially malignant disorder diagnosis. Oral Dis 2024; 30:801-804. [PMID: 35801664 DOI: 10.1111/odi.14304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/26/2022] [Accepted: 07/01/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Xi Yang
- Department of Oral and Maxillofacial Surgery, Zhang Zhiyuan Academician Workstation, Hainan Western Central Hospital, Shanghai Ninth People's Hospital, Danzhou, China
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanhuan Du
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Ji
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linjun Shi
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Liu
- College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Rivera-Peña B, Folawiyo O, Turaga N, Rodríguez-Benítez RJ, Felici ME, Aponte-Ortiz JA, Pirini F, Rodríguez-Torres S, Vázquez R, López R, Sidransky D, Guerrero-Preston R, Báez A. Promoter DNA methylation patterns in oral, laryngeal and oropharyngeal anatomical regions are associated with tumor differentiation, nodal involvement and survival. Oncol Lett 2024; 27:89. [PMID: 38268779 PMCID: PMC10804364 DOI: 10.3892/ol.2024.14223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/23/2023] [Indexed: 01/26/2024] Open
Abstract
Differentially methylated regions (DMRs) can be used as head and neck squamous cell carcinoma (HNSCC) diagnostic, prognostic and therapeutic targets in precision medicine workflows. DNA from 21 HNSCC and 10 healthy oral tissue samples was hybridized to a genome-wide tiling array to identify DMRs in a discovery cohort. Downstream analyses identified differences in promoter DNA methylation patterns in oral, laryngeal and oropharyngeal anatomical regions associated with tumor differentiation, nodal involvement and survival. Genome-wide DMR analysis showed 2,565 DMRs common to the three subsites. A total of 738 DMRs were unique to laryngeal cancer (n=7), 889 DMRs were unique to oral cavity cancer (n=10) and 363 DMRs were unique to pharyngeal cancer (n=6). Based on the genome-wide analysis and a Gene Ontology analysis, 10 candidate genes were selected to test for prognostic value and association with clinicopathological features. TIMP3 was associated with tumor differentiation in oral cavity cancer (P=0.039), DAPK1 was associated with nodal involvement in pharyngeal cancer (P=0.017) and PAX1 was associated with tumor differentiation in laryngeal cancer (P=0.040). A total of five candidate genes were selected, DAPK1, CDH1, PAX1, CALCA and TIMP3, for a prevalence study in a larger validation cohort: Oral cavity cancer samples (n=42), pharyngeal cancer tissues (n=25) and laryngeal cancer samples (n=52). PAX1 hypermethylation differed across HNSCC anatomic subsites (P=0.029), and was predominantly detected in laryngeal cancer. Kaplan-Meier survival analysis (P=0.043) and Cox regression analysis of overall survival (P=0.001) showed that DAPK1 methylation is associated with better prognosis in HNSCC. The findings of the present study showed that the HNSCC subsites oral cavity, pharynx and larynx display substantial differences in aberrant DNA methylation patterns, which may serve as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Bianca Rivera-Peña
- Department of Biology, University of Puerto Rico, San Juan 00925, Puerto Rico
- Department of Pharmacology, University of Puerto Rico School of Medicine, San Juan 00936, Puerto Rico
- Department of Otolaryngology-Head and Neck Surgery, University of Puerto Rico School of Medicine, San Juan 00936, Puerto Rico
| | - Oluwasina Folawiyo
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nitesh Turaga
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Rosa J. Rodríguez-Benítez
- Department of General Social Sciences, Faculty of Social Sciences, University of Puerto Rico, San Juan 00925, Puerto Rico
| | - Marcos E. Felici
- Oral Health Division, Puerto Rico Department of Health, San Juan 00927, Puerto Rico
| | - Jaime A. Aponte-Ortiz
- Department of General Surgery, University of Puerto Rico School of Medicine, San Juan 00936, Puerto Rico
| | - Francesca Pirini
- Biosciences Laboratory, IRCCS Instituto Romagnolo per lo Studio dei Tumori ‘Dino Amadori’, Meldola I-47014, Italy
| | | | - Roger Vázquez
- Department of Biology, University of Puerto Rico, San Juan 00925, Puerto Rico
| | - Ricardo López
- Department of Biology, University of Puerto Rico, San Juan 00925, Puerto Rico
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Rafael Guerrero-Preston
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Research and Development, LifeGene-Biomarks, San Juan 00909, Puerto Rico
| | - Adriana Báez
- Department of Pharmacology, University of Puerto Rico School of Medicine, San Juan 00936, Puerto Rico
- Department of Otolaryngology-Head and Neck Surgery, University of Puerto Rico School of Medicine, San Juan 00936, Puerto Rico
| |
Collapse
|
7
|
Gil-Martín E, Ramos E, López-Muñoz F, Egea J, Romero A. Potential of melatonin to reverse epigenetic aberrations in oral cancer: new findings. EXCLI JOURNAL 2023; 22:1280-1310. [PMID: 38234969 PMCID: PMC10792176 DOI: 10.17179/excli2023-6624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024]
Abstract
It is now an accepted principle that epigenetic alterations cause cellular dyshomeostasis and functional changes, both of which are essential for the initiation and completion of the tumor cycle. Oral carcinogenesis is no exception in this regard, as most of the tumors in the different subsites of the oral cavity arise from the cross-reaction between (epi)genetic inheritance and the huge challenge of environmental stressors. Currently, the biochemical machinery is put at the service of the tumor program, halting the cell cycle, triggering uncontrolled proliferation, driving angiogenesis and resistance to apoptosis, until the archetypes of the tumor phenotype are reached. Melatonin has the ability to dynamically affect the epigenetic code. It has become accepted that melatonin can reverse (epi)genetic aberrations present in oral and other cancers, suggesting the possibility of enhancing the oncostatic capacity of standard multimodal treatments by incorporating this indolamine as an adjuvant. First steps in this direction confirm the potential of melatonin as a countermeasure to mitigate the detrimental side effects of conventional first-line radiochemotherapy. This single effect could produce synergies of extraordinary clinical importance, allowing doses to be increased and treatments not to be interrupted, ultimately improving patients' quality of life and prognosis. Motivated by the urgency of improving the medical management of oral cancer, many authors advocate moving from in vitro and preclinical research, where the bulk of melatonin cancer research is concentrated, to systematic randomized clinical trials on large cohorts. Recognizing the challenge to improve the clinical management of cancer, our motivation is to encourage comprehensive and robust research to reveal the clinical potential of melatonin in oral cancer control. To improve the outcome and quality of life of patients with oral cancer, here we provide the latest evidence of the oncolytic activity that melatonin can achieve by manipulating epigenetic patterns in oronasopharyngeal tissue.
Collapse
Affiliation(s)
- Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, Camilo José Cela University of Madrid (UCJC), 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute, 28041 Madrid, Spain
| | - Javier Egea
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
8
|
Dholariya S, Singh RD, Patel KA. Melatonin: Emerging Player in the Management of Oral Cancer. Crit Rev Oncog 2023; 28:77-92. [PMID: 37830217 DOI: 10.1615/critrevoncog.2023048934] [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: 10/14/2023]
Abstract
Oral cancer (OC) has emerged as a major medical and social issue in many industrialized nations due to the high death rate. It is becoming increasingly common in people under the age of 45, although the underlying causes and mechanisms of this increase remain unclear. Melatonin, as a pleiotropic hormone, plays a pivotal role in a wide variety of cellular and physiological functions. Mounting evidence supports melatonin's ability to modify/influence oral carcinogenesis, help in the reduction of the incidence of OC, and increase chemo- and radiosensitivity. Despite its potential anti-carcinogenic effects, the precise function of melatonin in the management of OC is not well understood. This review summarizes the current knowledge regarding melatonin function in anti-carcinogenesis mechanisms for OC. In addition, clinical assessment and the potential therapeutic utility of melatonin in OC are discussed. This review will provide a basis for researchers to create new melatonin-based personalized medicines for treating and preventing OC.
Collapse
Affiliation(s)
- Sagar Dholariya
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Rajkot, Gujarat, India
| | - Ragini D Singh
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Rajkot, Gujarat, India
| | | |
Collapse
|
9
|
Gabusi A, Gissi DB, Grillini S, Stefanini M, Tarsitano A, Marchetti C, Foschini MP, Montebugnoli L, Morandi L. Shared epigenetic alterations between oral cancer and periodontitis: a preliminary study. Oral Dis 2022. [PMID: 35567390 DOI: 10.1111/odi.14251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION We recently developed a non-invasive sampling procedure for oral squamous cell carcinoma (OSCC) detection based on DNA methylation analysis of a panel of 13 genes. Oral cancer, as well as acute and chronic inflammatory diseases, may influence the methylation level of several genes in the oral cavity. In the present study, we evaluated the presence of periodontal disease(PD) and the methylation status using our 13-gene panel. METHODS Oral brushing specimens were collected from three different patient groups: 23 gingival OSCC patients, 15 patients affected by PD, and 15 healthy volunteers lacking evidence of PD. DNA methylation analysis was performed and each sample was determined to be positive or negative based on a predefined cut-off value. RESULTS Positive results were found for 23/23 OSCC patients, 3/15 PD patients and 0/15 samples from healthy volunteers. The GP1BB and MIR193 genes in the PD group exhibited mean methylation levels similar to OSCC patients. ZAP70 showed different methylation levels among three groups. CONCLUSION Preliminary data identified shared epigenetic alterations between PD and OSCC patients in two inflammatory genes(GP1BB and MIR193). This study may help identify potential links between the two diseases and serve as a starting point for future research focused on pathogenesis.
Collapse
Affiliation(s)
- Andrea Gabusi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Davide B Gissi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Sara Grillini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Martina Stefanini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Achille Tarsitano
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,Oral and Maxillo-facial Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna
| | - Claudio Marchetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,Oral and Maxillo-facial Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna
| | - Maria Pia Foschini
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Lucio Montebugnoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Luca Morandi
- Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
10
|
Chen Q, Chu L, Li X, Li H, Zhang Y, Cao Q, Zhuang Q. Investigation of an FGFR-Signaling-Related Prognostic Model and Immune Landscape in Head and Neck Squamous Cell Carcinoma. Front Cell Dev Biol 2022; 9:801715. [PMID: 35237609 PMCID: PMC8882630 DOI: 10.3389/fcell.2021.801715] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background: There is accumulating evidence on the clinical importance of the fibroblast growth factor receptor (FGFR) signal, hypoxia, and glycolysis in the immune microenvironment of head and neck squamous cell carcinoma (HNSCC), yet reliable prognostic signatures based on the combination of the fibrosis signal, hypoxia, and glycolysis have not been systematically investigated. Herein, we are committed to establish a fibrosis–hypoxia–glycolysis–related prediction model for the prognosis and related immune infiltration of HNSCC. Methods: Fibrotic signal status was estimated with microarray data of a discovery cohort from the TCGA database using the UMAP algorithm. Hypoxia, glycolysis, and immune-cell infiltration scores were imputed using the ssGSEA algorithm. Cox regression with the LASSO method was applied to define prognostic genes and develop a fibrosis–hypoxia–glycolysis–related gene signature. Immunohistochemistry (IHC) was conducted to identify the expression of specific genes in the prognostic model. Protein expression of several signature genes was evaluated in HPA. An independent cohort from the GEO database was used for external validation. Another scRNA-seq data set was used to clarify the related immune infiltration of HNSCC. Results: Six genes, including AREG, THBS1, SEMA3C, ANO1, IGHG2, and EPHX3, were identified to construct a prognostic model for risk stratification, which was mostly validated in the independent cohort. Multivariate analysis revealed that risk score calculated by our prognostic model was identified as an independent adverse prognostic factor (p < .001). Activated B cells, immature B cells, activated CD4+ T cells, activated CD8+ T cells, effector memory CD8+ T cells, MDSCs, and mast cells were identified as key immune cells between high- and low-risk groups. IHC results showed that the expression of SEMA3C, IGHG2 were slightly higher in HNSCC tissue than normal head and neck squamous cell tissue. THBS1, ANO1, and EPHX3 were verified by IHC in HPA. By using single-cell analysis, FGFR-related genes and highly expressed DEGs in low-survival patients were more active in monocytes than in other immune cells. Conclusion: A fibrosis–hypoxia–glycolysis–related prediction model provides risk estimation for better prognoses to patients diagnosed with HNSCC.
Collapse
Affiliation(s)
- Qi Chen
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ling Chu
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Li
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ying Zhang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingtai Cao
- Hunan Normal University School of Medicine, Changsha, China
| | - Quan Zhuang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, China
- *Correspondence: Quan Zhuang,
| |
Collapse
|
11
|
Jafarpour S, Saberi F, Yazdi M, Nedaeinia R, Amini G, Ferns GA, Salehi R. Association between colorectal cancer and the degree of ITGA4 promoter methylation in peripheral blood mononuclear cells. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Rossi R, Gissi DB, Gabusi A, Fabbri VP, Balbi T, Tarsitano A, Morandi L. A 13-Gene DNA Methylation Analysis Using Oral Brushing Specimens as an Indicator of Oral Cancer Risk: A Descriptive Case Report. Diagnostics (Basel) 2022; 12:diagnostics12020284. [PMID: 35204376 PMCID: PMC8870863 DOI: 10.3390/diagnostics12020284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Analysis of genetic or epigenetic markers from saliva or brushing specimens has been proposed as a diagnostic aid to identify patients at risk of developing oral cancer. However, no reliable non-invasive molecular method for this purpose is commercially available. In the present report, we describe the potential application of a procedure based on a 13-gene DNA methylation analysis using oral brushing samples from a patient affected by oral leukoplakia who developed two metachronous oral carcinomas during the follow-up period. A positive or a negative score was calculated for each brushing sample based on a predefined cut-off value. In this patient, a positive score was detected in the oral leukoplakia diagnosed more than 2 years before the development of oral squamous cell carcinoma and subsequently in clinically healthy mucosa 8 months before the appearance of a secondary tumor. This suggests a potential role of our procedure as an indicator of oral cancer risk.
Collapse
Affiliation(s)
- Roberto Rossi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy; (R.R.); (A.G.)
| | - Davide B. Gissi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy; (R.R.); (A.G.)
- Correspondence: ; Tel.: +39-05-1208-8123 (ext. 40125)
| | - Andrea Gabusi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40125 Bologna, Italy; (R.R.); (A.G.)
| | - Viscardo Paolo Fabbri
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology “M. Malpighi”, Bellaria Hospital, 40125 Bologna, Italy;
| | - Tiziana Balbi
- Unit of Anatomic Pathology S. Orsola Hospital, IRCCS Azienda Ospedaliero Universitaria, 40138 Bologna, Italy;
| | - Achille Tarsitano
- Maxillo-Facial Surgery Unit, Department of Biomedical and Neuromotor Sciences, IRCCS Azienda Ospedaliero Universitaria Bologna, University of Bologna, 40138 Bologna, Italy;
| | - Luca Morandi
- Functional and Molecular Neuroimaging Unit, Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy;
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| |
Collapse
|
13
|
Viet CT, Zhang X, Xu K, Yu G, Asam K, Thomas CM, Callahan NF, Doan C, Walker PC, Nguyen K, Kidd SC, Lee SC, Grandhi A, Allen CT, Young S, Melville JC, Shum JW, Viet DT, Herford AS, Roden DF, Gonzalez ML, Zhong JF, Aouizerat BE. Brush swab as a noninvasive surrogate for tissue biopsies in epigenomic profiling of oral cancer. Biomark Res 2021; 9:90. [PMID: 34930473 PMCID: PMC8686381 DOI: 10.1186/s40364-021-00349-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/28/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) has poor survival rates. There is a pressing need to develop more precise risk assessment methods to tailor clinical treatment. Epigenome-wide association studies in OSCC have not produced a viable biomarker. These studies have relied on methylation array platforms, which are limited in their ability to profile the methylome. In this study, we use MethylCap-Seq (MC-Seq), a comprehensive methylation quantification technique, and brush swab samples, to develop a noninvasive, readily translatable approach to profile the methylome in OSCC patients. METHODS Three OSCC patients underwent collection of cancer and contralateral normal tissue and brush swab biopsies, totaling 4 samples for each patient. Epigenome-wide DNA methylation quantification was performed using the SureSelectXT Methyl-Seq platform. DNA quality and methylation site resolution were compared between brush swab and tissue samples. Correlation and methylation value difference were determined for brush swabs vs. tissues for each respective patient and site (i.e., cancer or normal). Correlations were calculated between cancer and normal tissues and brush swab samples for each patient to determine the robustness of DNA methylation marks using brush swabs in clinical biomarker studies. RESULTS There were no significant differences in DNA yield between tissue and brush swab samples. Mapping efficiency exceeded 90% across all samples, with no differences between tissue and brush swabs. The average number of CpG sites with at least 10x depth of coverage was 2,716,674 for brush swabs and 2,903,261 for tissues. Matched tissue and brush swabs had excellent correlation (r = 0.913 for cancer samples and r = 0.951 for normal samples). The methylation profile of the top 1000 CpGs was significantly different between cancer and normal samples (mean p-value = 0.00021) but not different between tissues and brush swabs (mean p-value = 0.11). CONCLUSIONS Our results demonstrate that MC-Seq is an efficient platform for epigenome profiling in cancer biomarker studies, with broader methylome coverage than array-based platforms. Brush swab biopsy provides adequate DNA yield for MC-Seq, and taken together, our findings set the stage for development of a non-invasive methylome quantification technique for oral cancer with high translational potential.
Collapse
Affiliation(s)
- Chi T Viet
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, CA, USA.
| | - Xinyu Zhang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Ke Xu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Gary Yu
- New York University Rory Meyers College of Nursing, New York, NY, USA
| | - Kesava Asam
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, USA
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY, USA
| | - Carissa M Thomas
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas F Callahan
- Department of Oral and Maxillofacial Surgery, University of Illinois Chicago, College of Dentistry, Chicago, IL, USA
| | - Coleen Doan
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, CA, USA
| | - Paul C Walker
- Department of Otolaryngology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Khanh Nguyen
- Department of Otolaryngology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Stephanie C Kidd
- Department of Otolaryngology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Steve C Lee
- Department of Otolaryngology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Anupama Grandhi
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, CA, USA
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Simon Young
- Department of Oral, Head and Neck Oncology and Microvascular Reconstructive Surgery, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - James C Melville
- Department of Oral, Head and Neck Oncology and Microvascular Reconstructive Surgery, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jonathan W Shum
- Department of Oral, Head and Neck Oncology and Microvascular Reconstructive Surgery, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Alan S Herford
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, CA, USA
| | - Dylan F Roden
- Department of Otolaryngology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Manuel L Gonzalez
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jiang F Zhong
- Department of Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Bradley E Aouizerat
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
- New York University Rory Meyers College of Nursing, New York, NY, USA
| |
Collapse
|
14
|
Ricci C, Morandi L, Ambrosi F, Righi A, Gibertoni D, Maletta F, Agostinelli C, Corradini AG, Uccella S, Asioli S, Sessa F, La Rosa S, Papotti MG, Asioli S. Intron 4-5 hTERT DNA Hypermethylation in Merkel Cell Carcinoma: Frequency, Association with Other Clinico-pathological Features and Prognostic Relevance. Endocr Pathol 2021; 32:385-395. [PMID: 33909215 PMCID: PMC8370894 DOI: 10.1007/s12022-021-09669-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/23/2022]
Abstract
Merkel cell carcinoma (MCC) is an aggressive skin tumor with neuroendocrine differentiation, mainly affecting elderly population or immunocompromised individuals. As methylation of the human telomerase reverse transcriptase (mhTERT) has been shown to be a prognostic factor in different tumors, we investigated its role in MCC, in particular in intron 4-5 where rs10069690 has been mapped and recognized as a cancer susceptibility locus. DNA methylation analysis of hTERT gene was assessed retrospectively in a cohort of 69 MCC patients from the University of Bologna, University of Turin and University of Insubria. Overall mortality was evaluated with Kaplan-Meier curves and multivariable Royston-Parmar models. High levels of mhTERT (mhTERThigh) (HR = 2.500, p = 0.015) and p63 (HR = 2.659, p = 0.016) were the only two clinico-pathological features significantly associated with a higher overall mortality at the multivariate analysis. We did not find different levels of mhTERT between MCPyV (+) and (-) cases (21 vs 14, p = 0.554); furthermore, mhTERThigh was strongly associated with older age (80.5 vs 72 years, p = 0.026), no angioinvasion (40.7% vs 71.0%, p = 0.015), lower Ki67 (50 vs 70%, p = 0.005), and PD-L1 expressions in both tumor (0 vs 3%, p = 0.021) and immune cells (0 vs 10%, p = 0.002). mhTERT is a frequently involved epigenetic mechanism and a relevant prognostic factor in MCC. In addition, it belongs to the shared oncogenic pathways of MCC (MCPyV and UV-radiations) and it could be crucial, together with other epigenetic and genetic mechanisms as gene amplification, in determining the final levels of hTERT mRNA and telomerase activity in these patients.
Collapse
Affiliation(s)
- Costantino Ricci
- Pathology Unit, Maggiore Hospital, AUSL Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | | | - Alberto Righi
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Dino Gibertoni
- Department of Biomedical and Neuromotor Sciences, Unit of Hygiene and Biostatistics, University of Bologna, Bologna, Italy
| | - Francesca Maletta
- Department of Oncology, University of Turin, Città Della Salute Hospital, Turin, Italy
| | - Claudio Agostinelli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Angelo Gianluca Corradini
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | - Silvia Uccella
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Silvia Asioli
- Unit of Pathology, Morgagni-Pierantoni Hospital, Forlì, 47121, Italy
| | - Fausto Sessa
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Stefano La Rosa
- Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mauro Giulio Papotti
- Department of Oncology, University of Turin, Città Della Salute Hospital, Turin, Italy
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM) Surgical Pathology Section- Alma Mater Studiorum , University of Bologna , Bologna, Italy.
| |
Collapse
|
15
|
Maresca A, Del Dotto V, Capristo M, Scimonelli E, Tagliavini F, Morandi L, Tropeano CV, Caporali L, Mohamed S, Roberti M, Scandiffio L, Zaffagnini M, Rossi J, Cappelletti M, Musiani F, Contin M, Riva R, Liguori R, Pizza F, La Morgia C, Antelmi E, Loguercio Polosa P, Mignot E, Zanna C, Plazzi G, Carelli V. DNMT1 mutations leading to neurodegeneration paradoxically reflect on mitochondrial metabolism. Hum Mol Genet 2021; 29:1864-1881. [PMID: 31984424 PMCID: PMC7372549 DOI: 10.1093/hmg/ddaa014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
ADCA-DN and HSN-IE are rare neurodegenerative syndromes caused by dominant mutations in the replication foci targeting sequence (RFTS) of the DNA methyltransferase 1 (DNMT1) gene. Both phenotypes resemble mitochondrial disorders, and mitochondrial dysfunction was first observed in ADCA-DN. To explore mitochondrial involvement, we studied the effects of DNMT1 mutations in fibroblasts from four ADCA-DN and two HSN-IE patients. We documented impaired activity of purified DNMT1 mutant proteins, which in fibroblasts results in increased DNMT1 amount. We demonstrated that DNMT1 is not localized within mitochondria, but it is associated with the mitochondrial outer membrane. Concordantly, mitochondrial DNA failed to show meaningful CpG methylation. Strikingly, we found activated mitobiogenesis and OXPHOS with significant increase of H2O2, sharply contrasting with a reduced ATP content. Metabolomics profiling of mutant cells highlighted purine, arginine/urea cycle and glutamate metabolisms as the most consistently altered pathways, similar to primary mitochondrial diseases. The most severe mutations showed activation of energy shortage AMPK-dependent sensing, leading to mTORC1 inhibition. We propose that DNMT1 RFTS mutations deregulate metabolism lowering ATP levels, as a result of increased purine catabolism and urea cycle pathways. This is associated with a paradoxical mitochondrial hyper-function and increased oxidative stress, possibly resulting in neurodegeneration in non-dividing cells.
Collapse
Affiliation(s)
- Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Mariantonietta Capristo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Emanuela Scimonelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Francesca Tagliavini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | | | - Leonardo Caporali
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Susan Mohamed
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Marina Roberti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Letizia Scandiffio
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Mirko Zaffagnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Jacopo Rossi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Martina Cappelletti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Francesco Musiani
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Manuela Contin
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Roberto Riva
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Fabio Pizza
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Elena Antelmi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Paola Loguercio Polosa
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Emmanuel Mignot
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94304, USA
| | - Claudia Zanna
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Giuseppe Plazzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| |
Collapse
|
16
|
Lee EY, Song JM, Kim HJ, Park HR. Hypomethylation of lncRNA H19 as a potential prognostic biomarker for oral squamous cell carcinoma. Arch Oral Biol 2021; 129:105214. [PMID: 34333230 DOI: 10.1016/j.archoralbio.2021.105214] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Dysregulated DNA methylation is common in cancers and is considered one of the most important triggers in cancer development and progression. The expression and promoter methylation status of long non-coding RNA (lncRNA) H19 play a key role in several cancers, but its role is unclear in oral cancer. The aim of this study was to evaluate the potential of lncRNA H19 as a prognostic biomarker for oral cancer. DESIGNS The transcript levels and the methylation status of lncRNA H19 in OSCC cell lines and OSCC patient tissues were investigated by quantitative real-time RT-PCR (qRT-PCR) and methylation-specific PCR (MSP). Methylation ratio (%) were calculated from the intensity of the MSP in the gel image and Kaplan-Meier survival analysis of OSCC patient survival was performed for patients grouped according to the lncRNA H19 promoter methylation ratio. RESULTS lncRNA H19 was highly expressed and its promoter region was hypomethylated in OSSC cell lines as compared to normal control. Almost all OSCC patients tissues (63 out of 65, 97 %) showed hypomethylation of lncRNA H19 compared to normal oral mucosa tissues. There was a significant correlation between methylation ratio and tumor histopathologic grade. OSCC patients with hypomethylation of lncRNA H19 had a significantly lower 5-year survival rate. CONCLUSIONS Hypomethylation of lncRNA H19 may serve as a potential prognostic biomarker for oral cancer.
Collapse
Affiliation(s)
- Eun Young Lee
- Department of Oral Pathology, and BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea; Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jae Min Song
- Department of Oral and Maxillofacial Surgery, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea
| | - Hye Jung Kim
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea.
| | - Hae Ryoun Park
- Department of Oral Pathology, and BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea; Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Gyeongsangnam-do, Republic of Korea.
| |
Collapse
|
17
|
Pierik AS, Leemans CR, Brakenhoff RH. Resection Margins in Head and Neck Cancer Surgery: An Update of Residual Disease and Field Cancerization. Cancers (Basel) 2021; 13:2635. [PMID: 34071997 PMCID: PMC8198309 DOI: 10.3390/cancers13112635] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
Surgery is one of the mainstays of head and neck cancer treatment, and aims at radical resection of the tumor with 1 cm tumor-free margins to obtain locoregional control. Surgical margins are evaluated by histopathological examination of the resection specimen. It has been long an enigma that approximately 10-30% of surgically treated head and neck cancer patients develop locoregional recurrences even though the resection margins were microscopically tumor-free. However, the origins of these recurrences have been elucidated by a variety of molecular studies. Recurrences arise either from minimal residual disease, cancer cells in the surgical margins that escape detection by the pathologist when examining the specimen, or from precancerous mucosal changes that may remain unnoticed. Head and neck tumors develop in mucosal precursor changes that are sometimes visible but mostly not, fueling research into imaging modalities such as autofluorescence, to improve visualization. Mostly unnoticed, these precancerous changes may stay behind when the tumor is resected, and subsequent malignant progression will cause a local relapse. This led to a clinical trial of autofluorescence-guided surgery, of which the results were reported in 2020. This review focuses on the most recent literature of the improved diagnosis of the resection margins of surgically treated head and neck cancer patients, the pathobiological origin of recurrent disease, and relevant biomarkers to predict local relapse. Directions for further research will be discussed, including potential options for improved and personalized treatment, based on the most recently published data.
Collapse
Affiliation(s)
| | | | - Ruud H. Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam Tumor Biology and Immunology Section, Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.S.P.); (C.R.L.)
| |
Collapse
|
18
|
Gissi DB, Gabusi A, Tarsitano A, Asioli S, Rossi R, Marchetti C, Montebugnoli L, Foschini MP, Morandi L. Application of a non-invasive oral brushing procedure based on bisulfite sequencing of a 13-gene panel to study high-risk OSCC patients. Cancer Biomark 2021; 28:499-510. [PMID: 32568174 DOI: 10.3233/cbm-190422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND A non-invasive sampling procedure for the early detection of Oral Squamous Cell Carcinoma (OSCC) based on DNA methylation analysis of a panel of 13 genes was applied in 4 different OSCC risk-group of patients. Aim of the study is to evaluate the between-group differences and the variables related to the methylation profile of each group. METHODS Oral brushing samples were collected from 54 healthy subjects, 31 Oral Leukoplakia (OL) patients, 18 Oral Lichen Planus (OLP) patients and 26 patients previously treated for OSCC. Each sample was considered positive or negative in relation to a predefined cut-off value. RESULTS None of the samples from 54 healthy subjects were positive, whereas 22/31 OL, 3/18 OLP and 8/26 surgically treated OSCC samples showed positive values with respect to the cut-off. In OL patients, dysplasia was the only variable significantly related to positive values: 10/10 OLs with high-grade dysplasia were positive with respect to 12/21 OLs without dysplasia (Chi 6.039, p< 0.05). CONCLUSION DNA methylation analysis in epithelial cells collected by oral brushing seems to be a promising genetic method to distinguish lesions at high risk of developing OSCC. Larger population studies and an adequate follow-up period are necessary to confirm these preliminary data.
Collapse
Affiliation(s)
- Davide B Gissi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Andrea Gabusi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Achille Tarsitano
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Section of Maxillo-facial Surgery at Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology at Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Roberto Rossi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Claudio Marchetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Section of Maxillo-facial Surgery at Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Lucio Montebugnoli
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Maria P Foschini
- Department of Biomedical and Neuromotor Sciences, Section of Anatomic Pathology at Bellaria Hospital, University of Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto Delle Scienze Neurologiche di Bologna, University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Zhang H, Zhang Z, Liu X, Duan H, Xiang T, He Q, Su Z, Wu H, Liang Z. DNA Methylation Haplotype Block Markers Efficiently Discriminate Follicular Thyroid Carcinoma from Follicular Adenoma. J Clin Endocrinol Metab 2021; 106:1011-1021. [PMID: 33394038 PMCID: PMC7993581 DOI: 10.1210/clinem/dgaa950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Indexed: 12/19/2022]
Abstract
CONTEXT Follicular thyroid carcinoma (FTC) is the second most common type of thyroid carcinoma and must be pathologically distinguished from benign follicular adenoma (FA). Additionally, the clinical assessment of thyroid tumors with uncertain malignant potential (TT-UMP) demands effective indicators. OBJECTIVE We aimed to identify discriminating DNA methylation markers between FA and FTC. METHODS DNA methylation patterns were investigated in 33 FTC and 33 FA samples using reduced representation bisulfite sequencing and methylation haplotype block-based analysis. A prediction model was constructed and validated in an independent cohort of 13 FTC and 13 FA samples. Moreover, 36 TT-UMP samples were assessed using this model. RESULTS A total of 70 DNA methylation markers, approximately half of which were located within promoters, were identified to be significantly different between the FTC and FA samples. All the Gene Ontology terms enriched among the marker-associated genes were related to "DNA binding," implying that the inactivation of DNA binding played a role in FTC development. A random forest model with an area under the curve of 0.994 was constructed using those markers for discriminating FTC from FA in the validation cohort. When the TT-UMP samples were scored using this model, those with fewer driver mutations also exhibited lower scores. CONCLUSION An FTC-predicting model was constructed using DNA methylation markers, which distinguished between FA and FTC tissues with a high degree of accuracy. This model can also be used to help determine the potential of malignancy in TT-UMP.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | | | - Xiaoding Liu
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Huanli Duan
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | | - Qiye He
- Singlera Genomics Inc. Shanghai, China
| | - Zhixi Su
- Singlera Genomics Inc. Shanghai, China
| | - Huanwen Wu
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Correspondence: Zhiyong Liang, PhD, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China. ; or Huanwen Wu, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China.
| | - Zhiyong Liang
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Correspondence: Zhiyong Liang, PhD, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China. ; or Huanwen Wu, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
20
|
Renzi A, Morandi L, Bellei E, Marconato L, Rigillo A, Aralla M, Lenzi J, Bettini G, Tinto D, Sabattini S. Validation of oral brushing as a non-invasive technique for the identification of feline oral squamous cell carcinoma by DNA methylation and TP53 mutation analysis. Vet Comp Oncol 2021; 19:501-509. [PMID: 33624422 DOI: 10.1111/vco.12688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 02/13/2021] [Indexed: 11/27/2022]
Abstract
Feline oral squamous cell carcinoma (FOSCC) is a frequent and progressively invasive tumour. Early lesions are difficult to recognize based on the sole clinical examination and may be misinterpreted as non-neoplastic. Mutations of TP53 and epigenetic alterations of specific genes are present in FOSCC and may be early detected. Aim of this prospective study was to investigate the DNA methylation pattern of a 17-gene panel and TP53 mutational status of FOSCC cytological samples obtained by oral brushing. Results were compared with a control group, in order to validate this non-invasive procedure for the screening of FOSCC. In FOSCC, the same analyses were carried out on the corresponding histological sample, if available. Thirty-five FOSCC and 60 controls were included. Mutations of TP53 were detected in 17 FOSCC brushings (48%) and in none of the controls (P < .001). Six genes (ZAP70, FLI1, MiR124-1, KIF1A, MAGEC2 and MiR363) were differentially methylated in FOSCC and were included in a methylation score. An algorithm based on TP53 mutational status and methylation score allowed to differentiate FOSCC from controls with a 69% sensitivity and a 97% specificity (accuracy, 86%). In 19 FOSCC histological samples, TP53 mutational status was fully concordant with brushings and a positive methylation score was observed in all cases. These results are promising for the identification of FOSCC by oral brushing, although some factors may limit the accuracy of this technique and further studies are required to assess its reproducibility in clinical practice.
Collapse
Affiliation(s)
- Andrea Renzi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Emma Bellei
- Ospedale Veterinario "I Portoni Rossi", Bologna, Italy
| | - Laura Marconato
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Antonella Rigillo
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | | - Jacopo Lenzi
- Department of Biomedical and Neuromotor Sciences, Section of Hygiene, Public Health and Medical Statistics, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuliano Bettini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Debora Tinto
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
21
|
Herreros-Pomares A, Llorens C, Soriano B, Bagan L, Moreno A, Calabuig-Fariñas S, Jantus-Lewintre E, Bagan J. Differentially methylated genes in proliferative verrucous leukoplakia reveal potential malignant biomarkers for oral squamous cell carcinoma. Oral Oncol 2021; 116:105191. [PMID: 33657465 DOI: 10.1016/j.oraloncology.2021.105191] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To explore the pathophysiology of proliferative verrucous leucoplakia (PVL) through a methylated DNA immunoprecipitation and high-throughput sequencing (MeDIP-seq) case-control study. MATERIALS AND METHODS Oral biopsies from ten PVL patients and five healthy individuals were obtained and used to compare their epigenetic patterns. Network biology methods and integrative analyses of MeDIP-seq and RNAseq data were applied to investigate functional relations among differentially methylated genes (DMGs). The value of selected genes as malignant biomarkers was evaluated in a large cohort of oral squamous cell carcinoma (OSCC) patients from TCGA. RESULTS A total of 4647 differentially methylated regions were found, with a prominent state of hypermethylation in PVL patients. At the gene level, differentially methylated regions (DMRs) covered 826 genes with distinct roles, including transcription factors and binding proteins with functions in cell adhesion, migration, proliferation, regulation of transcription, bone morphogenesis, and cell signalling. Network analysis revealed three major hubs, two of them collecting proteins related to the response of the patients to PVL and treatment and one hub collecting proteins related to PVL and cancer. The integrative analysis revealed 8 genes (ARTN, CD8A, GATA3, HOXD10, MYO7A, OSR2, PLCB1, and SPOCK2) significantly upregulated in PVL compared to control and 5 genes (ANKRD6, DLG2, GPX3, PITX2, and ZNF736) significantly downregulated. The status of de-regulation found for PVL patients was concordant with what was found for OSCC samples compared to normal adjacent tissue. CONCLUSION Our findings show the potential of methylation markers in PVL and suggest novel OSCC diagnostic biomarkers which may boost the development of novel epigenetic-based therapies.
Collapse
Affiliation(s)
- Alejandro Herreros-Pomares
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain; CIBERONC, Valencia, Spain
| | - Carlos Llorens
- Biotechvana, Parc Cientific, Universitat de València, Paterna, Valencia, Spain
| | - Beatriz Soriano
- Biotechvana, Parc Cientific, Universitat de València, Paterna, Valencia, Spain
| | - Leticia Bagan
- Medicina Oral Unit, Stomatology Department, Valencia University, Spain
| | - Andrea Moreno
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain; TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, Valencia, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain; CIBERONC, Valencia, Spain; TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, Valencia, Spain; Department of Pathology, Universitat de València, Valencia, Spain
| | - Eloísa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Valencia, Spain; CIBERONC, Valencia, Spain; TRIAL Mixed Unit, Centro de Investigación Príncipe Felipe-Fundación para la Investigación del Hospital General Universitario de Valencia, Valencia, Spain; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain.
| | - José Bagan
- CIBERONC, Valencia, Spain; Medicina Oral Unit, Stomatology Department, Valencia University, Spain; Department of Stomatology and Maxillofacial Surgery, Hospital General Universitario de Valencia, Valencia, Spain.
| |
Collapse
|
22
|
Gissi DB, Morandi L, Colella G, De Luca R, Campisi G, Mauceri R, Romeo U, Tenore G, Mignogna MD, Adamo D, Oteri G, Marcianò A, Vescovi P, Meleti M, Serpico R, Di Stasio D, Pentenero M, Santarelli A, Bianchi A, Crimi S, Marchetti C, Tarsitano A, Foschini MP, Montebugnoli L. Clinical validation of 13-gene DNA methylation analysis in oral brushing samples for detection of oral carcinoma: Italian multicenter study. Head Neck 2021; 43:1563-1573. [PMID: 33511711 DOI: 10.1002/hed.26624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/27/2020] [Accepted: 01/15/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The aim of this Italian multicenter study was to evaluate the diagnostic performance of a minimally invasive method for the detection of oral squamous cell carcinoma (OSCC) based on 13-gene DNA methylation analysis in oral brushing samples. METHODS Oral brushing specimens were collected in 11 oral medicine centers across Italy. Twenty brushing specimens were collected by each center, 10 from patients with OSCC, and 10 from healthy volunteers. DNA methylation analysis was performed in blindness, and each sample was determined as positive or negative based on a predefined cutoff value. RESULTS DNA amplification failed in 4 of 220 (1.8%) samples. Of the specimens derived from patients with OSCC, 93.6% (103/110) were detected as positive, and 84.9% (90/106) of the samples from healthy volunteers were negative. CONCLUSION These data confirmed the diagnostic performance of our novel procedure in a large cohort of brushing specimens collected from 11 different centers and analyzed in blindness.
Collapse
Affiliation(s)
- Davide B Gissi
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| | - Luca Morandi
- Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Colella
- Multidisciplinary Department of Medical-Surgical and Dental Specialities, University of Campania "L. Vanvitelli", Naples, Italy
| | - Roberto De Luca
- Multidisciplinary Department of Medical-Surgical and Dental Specialities, University of Campania "L. Vanvitelli", Naples, Italy
| | - Giuseppina Campisi
- Department of Surgical, Oncological, and oral Sciences, Sector of Oral Medicine "Valerio Margiotta", University of Palermo, Palermo, Italy
| | - Rodolfo Mauceri
- Department of Surgical, Oncological, and oral Sciences, Sector of Oral Medicine "Valerio Margiotta", University of Palermo, Palermo, Italy
| | - Umberto Romeo
- Department of Oral and Maxillofacial Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Gianluca Tenore
- Department of Oral and Maxillofacial Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Michele D Mignogna
- Oral Medicine Complex Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences - Head & Neck Clinical Section, "Federico II" University of Naples, Naples, Italy
| | - Daniela Adamo
- Oral Medicine Complex Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences - Head & Neck Clinical Section, "Federico II" University of Naples, Naples, Italy
| | - Giacomo Oteri
- Department of Biomedical, Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Antonia Marcianò
- Department of Biomedical, Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Paolo Vescovi
- Oral Medicine and Laser Surgery Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Marco Meleti
- Oral Medicine and Laser Surgery Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rosario Serpico
- Multidisciplinary Department of Medical-Surgical and Dental Specialities, University of Campania "L. Vanvitelli", Naples, Italy
| | - Dario Di Stasio
- Multidisciplinary Department of Medical-Surgical and Dental Specialities, University of Campania "L. Vanvitelli", Naples, Italy
| | - Monica Pentenero
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Turin, Turin, Italy
| | - Andrea Santarelli
- Department of Clinical Sciences and Stomatology, Marche Polytechnic University, Ancona, Italy
| | - Alberto Bianchi
- Section of Maxillo-Facial Surgery, Department of General Surgery, Policlinico - San Marco, University of Catania, Catania, Italy
| | - Salvatore Crimi
- Section of Maxillo-Facial Surgery, Department of General Surgery, Policlinico - San Marco, University of Catania, Catania, Italy
| | - Claudio Marchetti
- Section of Maxillo-Facial Surgery at Policlinico S. Orsola-Malpighi, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Achille Tarsitano
- Section of Maxillo-Facial Surgery at Policlinico S. Orsola-Malpighi, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Pia Foschini
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Lucio Montebugnoli
- Department of Biomedical and Neuromotor Sciences, Section of Oral Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Chen L, Wang D. Identification of potential CpG sites for oral squamous cell carcinoma diagnosis via integrated analysis of DNA methylation and gene expression. World J Surg Oncol 2021; 19:16. [PMID: 33468155 PMCID: PMC7816501 DOI: 10.1186/s12957-021-02129-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/11/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) accounts for more than 90% of the oral carcinomas and has a high fatality rate. This study aimed to identify potentially diagnostic biomarkers of OSCC through integrated analysis of DNA methylation and gene expression profiles. METHODS The DNA methylation profiles of OSCC patients from The Cancer Genome Atlas (TCGA) were analyzed to screen patients with CpG island methylator phenotype (CIMP) and investigate the relationship between CIMP and survival probability of OSCC patients. Differential methylation and expression analyses of the paired OSCC tumor and paracancerous samples from TCGA were performed. Logistic regression model was established, and the accuracy of this diagnostic model for OSCC was evaluated in validation sets from Gene Expression Omnibus (GEO). RESULTS OSCC patients with CIMP had lower survival probability than those without CIMP. The cg02860732 and cg04342955 were determined as candidate diagnostic methylation sites for OSCC. Logistic regression model was established based on cg02860732 and cg04342955 showed relatively high diagnostic accuracy in OSCC. CONCLUSIONS A diagnostic model for OSCC was identified based on the methylation sites cg02860732 and cg04342955, which might be favorable for the diagnosis of OSCC.
Collapse
Affiliation(s)
- Le Chen
- Department of Stomatology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road Intersection, Hexi District, Tianjin, 300211, China
| | - Dong Wang
- Department of Stomatology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road Intersection, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
24
|
Adeoye J, Alade AA, Zhu WY, Wang W, Choi SW, Thomson P. Efficacy of hypermethylated DNA biomarkers in saliva and oral swabs for oral cancer diagnosis: Systematic review and meta-analysis. Oral Dis 2021; 28:541-558. [PMID: 33423350 DOI: 10.1111/odi.13773] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVES This study aims to determine the diagnostic test accuracy (DTA) of hypermethylated DNA biomarkers in saliva and oral swabs for oral squamous cell carcinoma (OSCC) detection from the prevalidation studies available. MATERIALS AND METHODS Electronic database searching of PubMed, EMBASE, Cochrane Library, Scopus, Web of Science, and LILACS was conducted to identify relevant articles that were published between January 1, 2000, and August 1, 2020. RESULTS Meta-analysis was conducted based on 11 of 20 studies selected for review. Included studies had high bias concerns on the QUADAS-2 study assessment tool. We found that salivary and oral swab hypermethylation markers had better specificity than sensitivity for oral cancer detection. Summary sensitivity and specificity (95% CI) of hypermethylation panels were 86.2% (60-96.2) and 90.6% (85.9-93.9) while for individual markers, summary sensitivity and specificity (95% CI) were 70% (56.9-80.5) and 91.9% (80.3-96.9), respectively. Respective positive and negative likelihood ratios for combined markers were 9.2 (5.89-14.36) and 0.15 (0.05-0.5), and 8.61 (3.39-21.87) and 0.33 (0.22-0.49) for single-application biomarkers. CONCLUSION DNA hypermethylation biomarkers especially in combination have acceptable DTA that warrants further optimization with rigorous biomarker evaluation methods for conclusive determination of their efficacy.
Collapse
Affiliation(s)
- John Adeoye
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.,Oral Cancer Research Group, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Azeez Arisekola Alade
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, IA, USA.,Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Wang-Yong Zhu
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Weilan Wang
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.,Oral Cancer Research Group, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Siu-Wai Choi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.,Oral Cancer Research Group, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Peter Thomson
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.,Oral Cancer Research Group, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Di Dalmazi G, Morandi L, Rubin B, Pilon C, Asioli S, Vicennati V, De Leo A, Ambrosi F, Santini D, Pagotto U, Maffeis V, Fassina A, Fallo F. DNA Methylation of Steroidogenic Enzymes in Benign Adrenocortical Tumors: New Insights in Aldosterone-Producing Adenomas. J Clin Endocrinol Metab 2020; 105:5897139. [PMID: 32844182 DOI: 10.1210/clinem/dgaa585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/24/2020] [Indexed: 01/24/2023]
Abstract
CONTEXT DNA methylation has been identified among putative regulatory mechanisms for CYP11B2 expression in primary aldosteronism. OBJECTIVE The objective of this work is to investigate DNA methylation and expression of genes encoding steroidogenic enzymes in benign adrenocortical tumors. DESIGN AND SETTING This cross-sectional study took place at university hospitals. PATIENTS We collected fresh-frozen tissues from patients with benign adrenocortical adenomas (n = 48) (nonfunctioning n = 9, autonomous cortisol secretion n = 9, Cushing syndrome n = 17, aldosterone-producing [APA] n = 13) and adrenal cortex adjacent to APA (n = 12). We collected formalin-fixed, paraffin-embedded (FFPE) specimens of paired APA and concurrent aldosterone-producing cell clusters (APCCs) (n = 6). INTERVENTION DNA methylation levels were evaluated by quantitative bisulfite next-generation sequencing in fresh-frozen tissues (CYP11A1, CYP11B1, CYP11B2, CYP17A1, CYP21A2, HSD3B1, HSD3B2, NR5A1, STAR, and TSPO) and FFPE APA/APCC paired samples (CYP11B2). CYP11B1, CYP11B2, CYP17, CYP21, and STAR gene expressions were examined by quantitative real-time polymerase chain reaction. MAIN OUTCOME MEASURE The main outcome measure was DNA methylation. RESULTS CYP11B2 methylation levels were significantly lower in APA than in other adrenal tissues (P < .001). Methylation levels of the remaining genes were comparable among groups. Overall, CYP11B2 expression and DNA methylation were negatively correlated (ρ = -0.379; P = .003). In FFPE-paired APA/APCC samples, CYP11B2 methylation level was significantly lower in APA than in concurrent APCCs (P = .028). CONCLUSIONS DNA methylation plays a regulatory role for CYP11B2 expression and may contribute to aldosterone hypersecretion in APA. Lower CYP11B2 methylation levels in APA than in APCCs may suggest an APCC-to-APA switch via progressive CYP11B2 demethylation. Conversely, DNA methylation seems not to be relevant in regulating the expression of genes encoding steroidogenic enzymes other than CYP11B2.
Collapse
Affiliation(s)
- Guido Di Dalmazi
- Endocrinology and Diabetes Prevention and Care Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Italy
| | - Beatrice Rubin
- Clinica Medica 3, Department of Medicine, University of Padova, Italy
| | - Catia Pilon
- Clinica Medica 3, Department of Medicine, University of Padova, Italy
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Italy
| | - Valentina Vicennati
- Endocrinology and Diabetes Prevention and Care Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | | | | | | - Uberto Pagotto
- Endocrinology and Diabetes Prevention and Care Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Valeria Maffeis
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padova, Italy
| | - Ambrogio Fassina
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padova, Italy
| | - Francesco Fallo
- Clinica Medica 3, Department of Medicine, University of Padova, Italy
| |
Collapse
|
26
|
Gissi DB, Fabbri VP, Gabusi A, Lenzi J, Morandi L, Melotti S, Asioli S, Tarsitano A, Balbi T, Marchetti C, Montebugnoli L. Pre-Operative Evaluation of DNA Methylation Profile in Oral Squamous Cell Carcinoma Can Predict Tumor Aggressive Potential. Int J Mol Sci 2020; 21:ijms21186691. [PMID: 32937734 PMCID: PMC7555204 DOI: 10.3390/ijms21186691] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prognosis of oral squamous cell carcinoma (OSCC) is difficult to exactly assess on pre-operative biopsies. Since OSCC DNA methylation profile has proved to be a useful pre-operative diagnostic tool, the aim of the present study was to evaluate the prognostic impact of DNA methylation profile to discriminate OSCC with high and low aggressive potential. METHODS 36 OSCC cases underwent neoplastic cells collection by gentle brushing of the lesion, before performing a pre-operative biopsy. The CpG islands methylation status of 13 gene (ZAP70, ITGA4, KIF1A, PARP15, EPHX3, NTM, LRRTM1, FLI1, MiR193, LINC00599, MiR296, TERT, GP1BB) was studied by bisulfite Next Generation Sequencing (NGS). A Cox proportional hazards model via likelihood-based component-wise boosting was used to evaluate the prognostic power of the CpG sites. RESULTS The boosting estimation identified five CpGs with prognostic significance: EPHX3-24, EPHX3-26, ITGA4-3, ITGA4-4, and MiR193-3. The combination of significant CpGs provided promising results for adverse events prediction (Brier score = 0.080, C-index = 0.802 and AUC = 0.850). ITGA4 had a strong prognostic power in patients with early OSCC. CONCLUSIONS These data confirm that the study of methylation profile provides new insights into the molecular mechanisms of OSCC and can allow a better OSCC prognostic stratification even before surgery.
Collapse
Affiliation(s)
- Davide B. Gissi
- Section of Oral Science, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (D.B.G.); (A.G.); (L.M.)
| | - Viscardo P. Fabbri
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy; (V.P.F.); (S.M.); (S.A.)
| | - Andrea Gabusi
- Section of Oral Science, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (D.B.G.); (A.G.); (L.M.)
| | - Jacopo Lenzi
- Section of Hygiene, Public Health and Medical Statistics, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Luca Morandi
- Functional MR Unit, Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy
- Correspondence:
| | - Sofia Melotti
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy; (V.P.F.); (S.M.); (S.A.)
| | - Sofia Asioli
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy; (V.P.F.); (S.M.); (S.A.)
| | - Achille Tarsitano
- Unit of Oral and Maxillofacial Surgery, Azienda Ospedaliero-Universitaria di Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (A.T.); (C.M.)
| | - Tiziana Balbi
- Unit of Anatomic Pathology, S. Orsola Hospital, 40138 Bologna, Italy;
| | - Claudio Marchetti
- Unit of Oral and Maxillofacial Surgery, Azienda Ospedaliero-Universitaria di Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (A.T.); (C.M.)
| | - Lucio Montebugnoli
- Section of Oral Science, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (D.B.G.); (A.G.); (L.M.)
| |
Collapse
|
27
|
An Evolutionary Cancer Epigenetic Approach Revealed DNA Hypermethylation of Ultra-Conserved Non-Coding Elements in Squamous Cell Carcinoma of Different Mammalian Species. Cells 2020; 9:cells9092092. [PMID: 32933205 PMCID: PMC7565279 DOI: 10.3390/cells9092092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Ultra-conserved non-coding elements (UCNEs) are genomic sequences that exhibit > 95% sequence identity between humans, mammals, birds, reptiles, and fish. Recent findings reported their functional role in cancer. The aim of this study was to evaluate the DNA methylation modifications of UNCEs in squamous cell carcinoma (SCC) from different mammal species. Methods: Fifty SCCs from 26 humans, 17 cats, 3 dogs, 1 horse, 1 bovine, 1 badger, and 1 porcupine were investigated. Fourteen feline stomatitis and normal samples from 36 healthy human donors, 7 cats, 5 dogs, 5 horses, 2 bovines and 1 badger were collected as normal controls. Bisulfite next generation sequencing evaluated the DNA methylation level from seven UCNEs (uc.160, uc.283, uc.416, uc.339, uc.270, uc.299, and uc.328). Results: 57/59 CpGs were significantly different according to the Kruskal–Wallis test (p < 0.05) comparing normal samples with SCC. A common DNA hypermethylation pattern was observed in SCCs from all the species evaluated in this study, with an increasing trend of hypermethylation starting from normal mucosa, through stomatitis to SCC. Conclusions: Our findings indicate that UCNEs are hypermethylated in human SCC, and this behavior is also conserved among different species of mammals.
Collapse
|
28
|
Reis RSD, Santos JAD, Abreu PMD, Dettogni RS, Santos EDVWD, Stur E, Agostini LP, Anders QS, Alves LNR, Valle IBD, Lima MA, Souza ED, Podestá JRV, Zeidler SVV, Cordeiro-Silva MDF, Louro ID. Hypermethylation status of DAPK, MGMT and RUNX3 in HPV negative oral and oropharyngeal squamous cell carcinoma. Genet Mol Biol 2020; 43:e20190334. [PMID: 32870234 PMCID: PMC7452731 DOI: 10.1590/1678-4685-gmb-2019-0334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 06/28/2020] [Indexed: 01/08/2023] Open
Abstract
Squamous cell carcinoma of the oral cavity and oropharynx is the sixth most common type of cancer in the world. During tumorigenesis, gene promoter hypermethylation is considered an important mechanism of transcription silencing of tumor suppressor genes, such as DAPK, MGMT and RUNX3. These genes participate in signaling pathways related to apoptosis, DNA repair and proliferation whose loss of expression is possibly associated with cancer development and progression. In order to investigate associations between hypermethylation and clinicopathological and prognostic parameters, promoter methylation was evaluated in 72 HPV negative oral and oropharyngeal tumors using methylation-specific PCR. Hypermethylation frequencies found for DAPK, MGMT and RUNX3 were 38.88%, 19.44% and 1.38% respectively. Patients with MGMT hypermethylation had a better 2-year overall survival compared to patients without methylation. Being MGMT a repair gene for alkylating agents, it could be a biomarker of treatment response for patients who are candidates for cisplatin chemotherapy, predicting drug resistance. In view of the considerable levels of hypermethylation in cancer cells and, for MGMT, its prognostic relevance, DAPK and MGMT show potential as epigenetic markers, in a way that additional studies may test its viability and efficacy in clinical management.
Collapse
Affiliation(s)
- Raquel Silva Dos Reis
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | - Jéssica Aflávio Dos Santos
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | - Priscila Marinho de Abreu
- Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Biotecnologia, Vitória, ES, Brazil.,Universidade Federal do Espírito Santo, Departamento de Patologia, Laboratório de Patologia Molecular, Vitória, ES, Brazil
| | - Raquel Spinassé Dettogni
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | | | - Elaine Stur
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | - Lidiane Pignaton Agostini
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | - Quézia Silva Anders
- Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Ciências Fisiológicas, Vitória, ES, Brazil
| | - Lyvia Neves Rebello Alves
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil.,Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Biotecnologia, Vitória, ES, Brazil
| | - Isabella Bittencourt do Valle
- Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Biotecnologia, Vitória, ES, Brazil.,Universidade Federal do Espírito Santo, Departamento de Patologia, Laboratório de Patologia Molecular, Vitória, ES, Brazil
| | - Marília Arantes Lima
- Universidade Federal do Espírito Santo, Departamento de Patologia, Laboratório de Patologia Molecular, Vitória, ES, Brazil
| | - Evandro Duccini Souza
- Hospital Santa Rita de Cássia - SESA, Programa de Prevenção e Detecção Precoce do Câncer Bucal, Setor de Cirurgia de Cabeça e Pescoço, Vitória, ES, Brazil
| | - José Roberto Vasconcelos Podestá
- Hospital Santa Rita de Cássia - SESA, Programa de Prevenção e Detecção Precoce do Câncer Bucal, Setor de Cirurgia de Cabeça e Pescoço, Vitória, ES, Brazil
| | - Sandra Ventorin von Zeidler
- Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Biotecnologia, Vitória, ES, Brazil.,Universidade Federal do Espírito Santo, Departamento de Patologia, Laboratório de Patologia Molecular, Vitória, ES, Brazil
| | - Melissa de Freitas Cordeiro-Silva
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil
| | - Iúri Drumond Louro
- Universidade Federal do Espírito Santo, Departamento de Ciências Biológicas, Núcleo de Genética Humana e Molecular, Vitória, ES, Brazil.,Universidade Federal do Espírito Santo, Programa de Pós-Graduação em Biotecnologia, Vitória, ES, Brazil
| |
Collapse
|
29
|
Renzi A, Morandi L, Lenzi J, Rigillo A, Bettini G, Bellei E, Giacomini A, Tinto D, Sabattini S. Analysis of DNA methylation and TP53 mutational status for differentiating feline oral squamous cell carcinoma from non-neoplastic mucosa: A preliminary study. Vet Comp Oncol 2020; 18:825-837. [PMID: 32506786 DOI: 10.1111/vco.12624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Feline oral squamous cell carcinoma (FOSCC) is characterized by high local invasiveness and early bone lysis. The late diagnosis largely limits the efficacy of therapy and increases treatment-related morbidity. The aim of this exploratory study was to assess the methylation pattern of 10 candidate genes and TP53 mutational status in histologic samples of FOSCC. Results were compared with normal oral mucosa and oral inflammatory lesions, in order to establish a gene panel for FOSCC detection. For 10 cats, the above analyses were also performed on oral brushing samples, in order to explore the utility of these methods for screening purposes. Thirty-one FOSCC, 25 chronic inflammatory lesions and 12 controls were included. TP53 mutations were significantly more frequent in the FOSCC (68%) than in the non-neoplastic oral mucosa (3%; P <.001). Based on lasso regression analysis, a step-wise algorithm including TP53, FLI1, MiR124-1, KIF1A and MAGEC2 was proposed. The algorithm allowed to differentiate FOSCC with 94% sensitivity and 100% specificity (accuracy, 97%). When applying the proposed algorithm on 10 brushing samples, accuracy decreased to 80%. These results indicate that the altered DNA methylation of specific genes is present in FOSCC, together with a significant proportion of TP53 mutations. Such alterations are infrequent in normal oral mucosa and chronic stomatitis in cats, suggesting their involvement in feline oral carcinogenesis and their utility as diagnostic biomarkers. Further studies on a high number of brushing samples will be needed to assess the utility of a screening test for the early detection of FOSCC.
Collapse
Affiliation(s)
- Andrea Renzi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Jacopo Lenzi
- Department of Biomedical and Neuromotor Sciences, Section of Hygiene, Public Health and Medical Statistics, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Antonella Rigillo
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuliano Bettini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Emma Bellei
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Alessandra Giacomini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Debora Tinto
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
30
|
Xu Y, Zhao W, Mo Y, Ma N, Midorikawa K, Kobayashi H, Hiraku Y, Oikawa S, Zhang Z, Huang G, Takeuchi K, Murata M. Combination of RERG and ZNF671 methylation rates in circulating cell-free DNA: A novel biomarker for screening of nasopharyngeal carcinoma. Cancer Sci 2020; 111:2536-2545. [PMID: 32324312 PMCID: PMC7385361 DOI: 10.1111/cas.14431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/02/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in Southeast Asia, hence, identifying easily detectable biomarkers for NPC screening is essential for better diagnosis and prognosis. Using genome-wide and targeted analyses based on next-generation sequencing approaches, we previously showed that gene promoters are hypermethylated in NPC tissues. To confirm whether DNA methylation rates of genes could be used as biomarkers for NPC screening, 79 histologically diagnosed NPC patients and 29 noncancer patients were recruited. A convenient quantitative analysis of DNA methylation using real-time PCR (qAMP) was carried out, involving pretreatment of tissue DNA, and circulating cell-free DNA (ccfDNA) from nonhemolytic plasma, with methylation-sensitive and/or methylation-dependent restriction enzymes. The qAMP analyses revealed that methylation rates of RERG, ZNF671, ITGA4, and SHISA3 were significantly higher in NPC primary tumor tissues compared to noncancerous tissues, with sufficient diagnostic accuracy of the area under receiver operating characteristic curves (AUC). Interestingly, higher methylation rates of RERG in ccfDNA were statistically significant and yielded a very good AUC; however, those of ZNF671, ITGA4, and SHISA3 were not significant. Furthermore, the combination of methylation rates of RERG and ZNF671 in ccfDNA showed higher diagnostic accuracy than either of them individually. In conclusion, the methylation rates of specific genes in ccfDNA can serve as novel biomarkers for early detection and screening of NPC.
Collapse
Affiliation(s)
- Yifei Xu
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
- Department of Otorhinolaryngology – Head and Neck SurgeryMie University Graduate School of MedicineTsuJapan
- Department of Otorhinolaryngology – Head and Neck SurgeryFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Weilin Zhao
- Department of Otorhinolaryngology – Head and Neck SurgeryFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Yingxi Mo
- Department of ResearchAffiliated Tumor Hospital of Guangxi Medical UniversityNanningChina
| | - Ning Ma
- Graduate School of Health ScienceSuzuka University of Medical ScienceSuzukaJapan
| | - Kaoru Midorikawa
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
| | - Yusuke Hiraku
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
- Department of Environmental HealthUniversity of Fukui School of Medical ScienceEiheijiJapan
| | - Shinji Oikawa
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
| | - Zhe Zhang
- Department of Otorhinolaryngology – Head and Neck SurgeryFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Guangwu Huang
- Department of Otorhinolaryngology – Head and Neck SurgeryFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology – Head and Neck SurgeryMie University Graduate School of MedicineTsuJapan
| | - Mariko Murata
- Department of Environmental and Molecular MedicineMie University Graduate School of MedicineTsuJapan
| |
Collapse
|
31
|
Foschini MP, Morandi L, Sanchez AM, Santoro A, Mulè A, Zannoni GF, Varga Z, Moskovszky L, Cucchi MC, Moelans CB, Giove G, van Diest PJ, Masetti R. Methylation Profile of X-Chromosome-Related Genes in Male Breast Cancer. Front Oncol 2020; 10:784. [PMID: 32626651 PMCID: PMC7313421 DOI: 10.3389/fonc.2020.00784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Androgen receptor (AR) has been described to play a prominent role in male breast cancer (MBC). It maps on chromosome X, and recent reports indicate that X-chromosome polysomy is frequent in MBC. Since the response to anti-androgen therapy may depend on AR polysomy and on its overexpression similarly to prostate cancer, the aim of the present study was to investigate the DNA methylation level of AR and its coregulators, especially those mapped on the X-chromosome, that may influence the activity of AR in MBC. Methods: The DNA methylation level of AR, MAGEA2, MAGEA11, MAGEC1, MAGEC2, FLNA, HDAC6, and UXT, mapped on the X-chromosome, was evaluated by quantitative bisulfite-NGS. Bioinformatic analysis was performed in a Galaxy Project environment using BWA-METH, MethylDackel, and Methylation Plotter tools. The study population consisted of MBC (41 cases) compared with gynecomastia (17 cases). Results:MAGEA family members, especially MAGEA2, MAGEA11, MAGEC, and UXT and HDAC6 showed hypomethylation of several CpGs, reaching statistical significance by the Kruskal–Wallis test (p < 0.01) in MBC when compared to gynecomastia. AR showed almost no methylation at all. Conclusions: Our study demonstrated for the first time that MAGEA family members mapped on the X-chromosome and coregulators of AR are hypomethylated in MBC. This may lead to their overexpression, enhancing AR activity.
Collapse
Affiliation(s)
- Maria P Foschini
- Anatomic Pathology Section "M. Malpighi", Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Morandi
- Functional MR Unit, Department of Biomedical and Neuromotor Sciences, IRCCS Istituto delle Scienze Neurologiche di Bologna, University of Bologna, Bologna, Italy
| | - Alejandro M Sanchez
- Dipartimento Scienze della Salute della donna e del Bambino e di Sanità Pubblica, Multidisciplinary Breast Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angela Santoro
- Pathology Unit, Dipartimento Scienze della Salute della donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonino Mulè
- Pathology Unit, Dipartimento Scienze della Salute della donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gian Franco Zannoni
- Pathology Unit, Dipartimento Scienze della Salute della donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Zsuzsanna Varga
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Linda Moskovszky
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Maria C Cucchi
- Unit of Breast Surgery, Department of Oncology, Bellaria Hospital, AUSL Bologna, Bologna, Italy
| | - Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Gianluca Giove
- Anatomic Pathology Section "M. Malpighi", Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Masetti
- Dipartimento Scienze della Salute della donna e del Bambino e di Sanità Pubblica, Multidisciplinary Breast Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
32
|
Sun R, Juan YC, Su YF, Zhang WB, Yu Y, Yang HY, Yu GY, Peng X. Hypermethylated PAX1 and ZNF582 genes in the tissue sample are associated with aggressive progression of oral squamous cell carcinoma. J Oral Pathol Med 2020; 49:751-760. [PMID: 32428271 DOI: 10.1111/jop.13035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/22/2020] [Accepted: 05/01/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND DNA methylation of paired box gene 1 (PAX1) and zinc finger 582 (ZNF582) is promising cancer biomarkers for oral squamous cell carcinoma detection. This study aims to investigate the correlation between PAX1 or ZNF582 methylation and the progression of oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS A total of 135 OSCC cases from Peking University School and Hospital of Stomatology were enrolled in this study. Tissue specimens were collected from the lesion site and corresponding adjacent normal site. The methylation level of these two genes was evaluated in primary and recurrent OSCC group. RESULTS Hypermethylation of PAX1 or ZNF582 was observed in lesion sites among primary and recurrent OSCC cases. In the lesion site of primary cases, promoter methylation was observed in T3/T4 (PAX1: P = .02; ZNF582: P = .01), stage III/IV (PAX1: P = .03; ZNF582: P = .01), and bone invasion cases (PAX1: P = .02; ZNF582: P = .047). In the subgroup analysis, the correlation between hypermethylation and OSCC severity remains significant with exposure to smoking/alcohol consumption. CONCLUSIONS Hypermethylated PAX1 and ZNF582 can sufficiently act as biomarkers to reflect the severity or progression of OSCC.
Collapse
Affiliation(s)
- Rui Sun
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yi-Chen Juan
- iStat Biomedical Co., Ltd, New Taipei Citys, Taiwan
| | - Yee-Fun Su
- iStat Biomedical Co., Ltd, New Taipei Citys, Taiwan
| | - Wen-Bo Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yao Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hong-Yu Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xin Peng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
33
|
Xu X, Dai Y, Feng L, Zhang H, Hu Y, Xu L, Zhu X, Jiang Y. Knockdown of Nav1.5 inhibits cell proliferation, migration and invasion via Wnt/β-catenin signaling pathway in oral squamous cell carcinoma. Acta Biochim Biophys Sin (Shanghai) 2020; 52:527-535. [PMID: 32400862 DOI: 10.1093/abbs/gmaa021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/14/2019] [Accepted: 03/06/2020] [Indexed: 12/19/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common type of malignant oral cancer that has a high recurrence rate. Voltage-gated sodium channel Nav1.5 was reported to be highly up-regulated in various types of cancers. However, the regulatory mechanism of Nav1.5 in cancers including OSCC still remains elusive. In this study, Nav1.5 was found to be highly expressed in OSCC tissues and cells. Through the analysis of clinical characteristics of patients, we found that the expression level of Nav1.5 was closely related to neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, tumor-node-metastasis stage, and lymph node metastasis. Moreover, we found that Nav1.5 mainly located on the cell membrane as well as cytoplasm and knockdown of Nav1.5 promoted cell apoptosis and decreased proliferation in OSCC. Transwell assay results showed that knockdown of Nav1.5 effectively suppressed the migration and invasion in OSCC. In addition, knockdown of Nav1.5 was found to inhibit the protein and mRNA expression levels of β-catenin, cyclin D1, and c-Myc in the Wnt/β-catenin signaling pathway. In summary, these results indicated that Nav1.5 may be involved in the progression of OSCC through the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiaoli Xu
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Yongzheng Dai
- Hefei School of Stomatology, Anhui Medical University, Hefei 230001, China
- Department of General Dentistry, Hefei Stomatological Hospital, Hefei 230001, China
| | - Linfei Feng
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hongli Zhang
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Yukun Hu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Le Xu
- Department of Stomatology, the Fourth Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| | - Xinwei Zhu
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
- Binhu Clinical Division, Anhui Stomatology Hospital Affiliated to Anhui Medical University, Hefei 230601, China
| | - Yong Jiang
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
- Department of Stomatology, the Fourth Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| |
Collapse
|
34
|
Dai Y, Lv Q, Qi T, Qu J, Ni H, Liao Y, Liu P, Qu Q. Identification of hub methylated-CpG sites and associated genes in oral squamous cell carcinoma. Cancer Med 2020; 9:3174-3187. [PMID: 32155325 PMCID: PMC7196066 DOI: 10.1002/cam4.2969] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/07/2020] [Accepted: 02/16/2020] [Indexed: 12/15/2022] Open
Abstract
To improve personalized diagnosis and prognosis for oral squamous cell carcinoma (OSCC) by identification of hub methylated‐CpG sites and associated genes, weighted gene comethylation network analysis (WGCNA) was performed to examine and identify hub modules and CpG sites correlated with OSCC. Here, WGCNA modeling yielded blue and brown comethylation modules that were significantly associated with OSCC status. Following screening of the differentially expressed genes (DEGs) from gene expression microarrays and differentially methylated‐CpG sites (DCGs), integrated multiomics analysis of the DEGs, DCGs, and hub CpG sites from the modules was performed to investigate their correlations. Expression levels of 16 CpG sites‐associated genes were negatively correlated with methylation patterns of promoter. Moreover, Kaplan‐Meier survival analysis of the hub CpG sites and associated genes was carried out using 2 public databases, MethSurv and GEPIA. Only 5 genes, ACTA1, ACTN2, OSR1, SYNGR1, and ZNF677, had significant overall survival using GEPIA. Hypermethylated‐CpG sites ACTN2‐cg21376883 and OSR1‐cg06509239 were found to be associated with poor survival by MethSurv. Methylation status of specific site and expression levels of associated genes were determined using clinical samples by quantitative methylation‐specific PCR and real‐time PCR. Pearson's correlation analysis showed that methylation levels of cg06509239 and cg18335068 were negatively related to OSR1 and ZNF677 expression levels, respectively. Our classification schema using multiomics analysis represents a screening framework for identification of hub CpG sites and associated genes.
Collapse
Affiliation(s)
- Yuxin Dai
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qiaoli Lv
- Department of Science and Education, Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Tingting Qi
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongli Ni
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongkang Liao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Peng Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
35
|
Asioli S, Zoli M, Guaraldi F, Sollini G, Bacci A, Gibertoni D, Ricci C, Morandi L, Pasquini E, Righi A, Mazzatenta D. Peculiar pathological, radiological and clinical features of skull‐base de‐differentiated chordomas. Results from a referral centre case–series and literature review. Histopathology 2020; 76:731-739. [DOI: 10.1111/his.14024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Sofia Asioli
- Section of Anatomic Pathology ‘M. Malpighi’ Bellaria Hospital BolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM) University of Bologna Bologna Italy
- Pituitary Unit Department of Biomedical and Neuromotor Sciences (DIBINEM) of Neurological Sciences of Bologna Center for the Diagnosis and Treatment of Hypothalamic and Pituitary Diseases Bologna Italy
| | - Matteo Zoli
- Department of Biomedical and Neuromotor Sciences (DIBINEM) University of Bologna Bologna Italy
- Pituitary Unit Department of Biomedical and Neuromotor Sciences (DIBINEM) of Neurological Sciences of Bologna Center for the Diagnosis and Treatment of Hypothalamic and Pituitary Diseases Bologna Italy
| | - Federica Guaraldi
- Department of Biomedical and Neuromotor Sciences (DIBINEM) University of Bologna Bologna Italy
- Pituitary Unit Department of Biomedical and Neuromotor Sciences (DIBINEM) of Neurological Sciences of Bologna Center for the Diagnosis and Treatment of Hypothalamic and Pituitary Diseases Bologna Italy
| | | | - Antonella Bacci
- Division of Neuroradiology IRCCS Institute of Neurological Sciences of Bologna Bologna Italy
| | - Dino Gibertoni
- Unit of Hygiene, Public Health and Biostatistics Department of Biomedical and Neuromotor Sciences University of Bologna Bologna Italy
| | - Costantino Ricci
- Section of Anatomic Pathology ‘M. Malpighi’ Bellaria Hospital BolognaItaly
| | - Luca Morandi
- Section of Anatomic Pathology ‘M. Malpighi’ Bellaria Hospital BolognaItaly
| | | | - Alberto Righi
- Service of Anatomic Pathology IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Diego Mazzatenta
- Department of Biomedical and Neuromotor Sciences (DIBINEM) University of Bologna Bologna Italy
- Pituitary Unit Department of Biomedical and Neuromotor Sciences (DIBINEM) of Neurological Sciences of Bologna Center for the Diagnosis and Treatment of Hypothalamic and Pituitary Diseases Bologna Italy
| |
Collapse
|
36
|
Zhang J, Chen GY, Wang F, Zhou G. MiR-29b interacts with IFN-γ and induces DNA hypomethylation in CD4+ T cells of oral lichen planus. Int J Biol Macromol 2020; 147:1248-1254. [DOI: 10.1016/j.ijbiomac.2019.09.252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 12/17/2022]
|
37
|
Wang C, Zhuang X, Xu J, Dai Z, Wu W, Zhang C, Lin S, Chen S, Lin H, Tang W. Variants of MIR137HG Genes are Associated with Liver Cancer Risk in Chinese Li Population. Onco Targets Ther 2020; 13:1809-1818. [PMID: 32184616 PMCID: PMC7053808 DOI: 10.2147/ott.s225669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Liver cancer (LC) is the sixth most common cancer and the second leading cause of cancer mortality worldwide, and its incidence rate is high in China. Methods In this study, we aimed to investigate the contribution of MIR137HG (MIR137 Host Gene) polymorphisms to LC risk in a case–control study with 432 LC patients and 430 healthy controls. A logistic recession model was used to evaluate the effects of candidate single nucleotide polymorphisms (SNPs) on LC risk. HaploReg v 4.1 database was conducted to predict the potential functionality of SNPs. Results The results revealed that rs17371457 and rs7554283 in the MIR137HG gene were correlated with an enhanced LC risk under the allele (P = 0.001 and P = 0.043, respectively) and genetic models (P < 0.05). When the sample was stratified by gender and age, statistically significant associations were found. Rs9440302, rs17371457 and rs7554283 were associated with an increased the risk of LC among individuals aged >55 years (P < 0.05); rs17371457 was related to higher LC risk in males (P < 0.05). Similarly, the haplotype AG constituted by rs12333983 and rs3735451 significantly increased LC risk in Chinese Li population (P = 0.043). Six SNPs distributed in MIR137HG were successfully predicted as regulatory SNPs with different biological functions. Conclusion Our research firstly showed that MIR137HG gene polymorphisms were implicated in LC susceptibility among Chinese Li population.
Collapse
Affiliation(s)
- Chaoying Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Xiaohong Zhuang
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Junnv Xu
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China.,Hainan Medical University, Haikou, Hainan 571199, People's Republic of China
| | - Zhisheng Dai
- Department of Medical Oncology, The Second People's Hospital of Hainan Province, Wuzhishan, Hainan 572200, People's Republic of China
| | - Weixiong Wu
- Intensive Care Medicine 1 District, The Second Affiliated Hospital of Hainan Medical College, Haikou, Hainan 570311, People's Republic of China
| | - Chengsheng Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Shu Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Sehong Chen
- Department of Medical Oncology, The Second People's Hospital of Hainan Province, Wuzhishan, Hainan 572200, People's Republic of China
| | - Haifeng Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| | - Wenjun Tang
- Department of Medical Oncology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, People's Republic of China
| |
Collapse
|
38
|
Zhang W, Shang S, Yang Y, Lu P, Wang T, Cui X, Tang X. Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma. Exp Ther Med 2020; 19:2963-2972. [PMID: 32256782 PMCID: PMC7086284 DOI: 10.3892/etm.2020.8554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a painful and fatal disease that undoubtedly remains a health care priority and offers significant therapeutic challenges. The significance of epigenetic modifications, including DNA methylation in tumor development, has gained the attention of researchers. Identifying DNA methylation-driven genes and investigating the mechanisms underlying the tumorigenesis of PAAD are of substantial importance for developing methods of physiological evaluation, treatment planning and prognostic prediction for PAAD. In the present study, a comprehensive analysis of DNA methylation and gene expression data from 188 clinical samples was performed to identify DNA methylation-driven genes in PAAD. In addition, the diagnostic and prognostic value of DNA methylation-driven genes was evaluated using receiver operating characteristic curve, survival and recurrence analyses. A total of 7 DNA methylation-driven genes, namely zinc finger protein 208 (ZNF208), eomesodermin (EOMES), prostaglandin D2 receptor (PTGDR), chromosome 12 open reading frame 42 (C12orf42), integrin subunit α 4 (ITGA4), dedicator of cytokinesis 8 and protein phosphatase 1 regulatory inhibitor subunit 14D (PPP1R14D), were identified. All of them may be used to diagnose PAAD with excellent specificity and sensitivity (area under curve, >0.8). Of the 7 DNA methylation-driven genes, 6 were significantly associated with overall survival (OS) and recurrence-free survival (RFS) P<0.05). Among them, ZNF208, EOMES, PTGDR, C12orf42 and ITGA4 were significantly negatively associated with the OS rate and positively associated with the recurrence rate, while PPP1R14D was significantly positively associated with the OS rate and negatively associated with the recurrence rate. The present study provides novel insight into the epigenetic alterations associated with the occurrence and progression of PAAD, thereby increasing the mechanistic understanding of this disease, offering potential novel molecular biomarkers and contributing to the development of therapeutic targets for PAAD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Shuai Shang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Yingying Yang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Peiyao Lu
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Teng Wang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Xinyi Cui
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Xuexi Tang
- Department of Marine Ecology, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, P.R. China.,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory of Oceanology for Marine Science and Technology, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
39
|
Gissi DB, Tarsitano A, Gabusi A, Rossi R, Attardo G, Lenzi J, Marchetti C, Montebugnoli L, Foschini MP, Morandi L. 13-gene DNA Methylation Analysis from Oral Brushing: A Promising Non Invasive Tool in the Follow-up of Oral Cancer Patients. J Clin Med 2019; 8:jcm8122107. [PMID: 31810211 PMCID: PMC6947392 DOI: 10.3390/jcm8122107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Background: This study aimed to evaluate the prognostic value of a non-invasive sampling procedure based on 13-gene DNA methylation analysis in the follow-up of patients previously treated for oral squamous cell carcinoma (OSCC). Methods: The study population included 49 consecutive patients treated for OSCC. Oral brushing sample collection was performed at two different times: before any cancer treatment in the tumor mass and during patient follow-up almost 6 months after OSCC treatment, within the regenerative area after OSCC resection. Each sample was considered positive or negative in relation to a predefined cut-off value. Results: Before any cancer treatment, 47/49 specimens exceeded the score and were considered as positive. Six months after OSCC resection, 16/49 specimens also had positive scores in the samples collected from the regenerative area. During the follow-up period, 7/49 patients developed locoregional relapse: 6/7 patients had a positive score in the regenerative area after OSCC resection. The presence of a positive score after oral cancer treatment was the most powerful variable related to the appearance of locoregional relapse. Conclusion: 13-gene DNA methylation analysis by oral brushing may have a clinical application as a prognostic non-invasive tool in the follow-up of patients surgically treated for OSCC.
Collapse
Affiliation(s)
- Davide B. Gissi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (A.G.); (R.R.); (L.M.)
- Correspondence: ; Tel.: +39-0512088123
| | - Achille Tarsitano
- Section of Maxillo-Facial Surgery at Policlinico S. Orsola-Malpighi, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (A.T.); (C.M.)
| | - Andrea Gabusi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (A.G.); (R.R.); (L.M.)
| | - Roberto Rossi
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (A.G.); (R.R.); (L.M.)
| | - Giuseppe Attardo
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy; (G.A.); (M.P.F.)
| | - Jacopo Lenzi
- Section of Hygiene, Department of Biomedical and Neuromotor Sciences, Public Health and Medical Statistics, University of Bologna, 40126 Bologna, Italy
| | - Claudio Marchetti
- Section of Maxillo-Facial Surgery at Policlinico S. Orsola-Malpighi, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (A.T.); (C.M.)
| | - Lucio Montebugnoli
- Section of Oral Sciences, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40159 Bologna, Italy; (A.G.); (R.R.); (L.M.)
| | - Maria P. Foschini
- Section of Anatomic Pathology at Bellaria Hospital, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy; (G.A.); (M.P.F.)
| | - Luca Morandi
- Functional MR Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy;
| |
Collapse
|
40
|
Bai G, Song J, Yuan Y, Chen Z, Tian Y, Yin X, Niu Y, Liu J. Systematic analysis of differentially methylated expressed genes and site-specific methylation as potential prognostic markers in head and neck cancer. J Cell Physiol 2019; 234:22687-22702. [PMID: 31131446 PMCID: PMC6772109 DOI: 10.1002/jcp.28835] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/27/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022]
Abstract
Head and neck cancer (HNC) remains one of the most malignant tumors with a significantly high mortality. DNA methylation exerts a vital role in the prognosis of HNC. In this study, we try to screen abnormal differential methylation genes (DMGs) and pathways in Head-Neck Squamous Cell Carcinoma via integral bioinformatics analysis. Data of gene expression microarrays and gene methylation microarrays were obtained from the Cancer Genome Atlas database. Aberrant DMGs were identified by the R Limma package. We conducted the Cox regression analysis to select the prognostic aberrant DMGs and site-specific methylation. Five aberrant DMGs were recognized that significantly correlated with overall survival. The prognostic model was constructed based on five DMGs (PAX9, STK33, GPR150, INSM1, and EPHX3). The five DMG models acted as prognostic biomarkers for HNC. The area under the curve based on the five DMGs predicting 5-year survival is 0.665. Moreover, the correlation between the DMGs/site-specific methylation and gene expression was also explored. The findings demonstrated that the five DMGs can be used as independent prognostic biomarkers for predicting the prognosis of patients with HNC. Our study might lay the groundwork for further mechanism exploration in HNC and may help identify diagnostic biomarkers for early stage HNC.
Collapse
Affiliation(s)
- Guohui Bai
- Zunyi Medical UniversityZunyiGuizhouChina
- Special Key Laboratory of Oral Diseases ResearchStomatological Hospital Affiliated to Zunyi Medical UniversityGuizhouChina
| | - Jukun Song
- Department of Oral and Maxillofacial SurgeryGuizhou Provincial People's HospitalGuizhouChina
| | | | - Zhu Chen
- Guiyang Hospital of StomatologyGuizhouChina
| | - Yuan Tian
- Stomatology Colledge Affiliated to Zunyi Medical UniversityZunyiGuizhouChina
| | - Xinhai Yin
- Department of Oral and Maxillofacial SurgeryGuizhou Provincial People's HospitalGuizhouChina
| | - Yuming Niu
- Department of Stomatology and Center for Evidence‐Based Medicine and Clinical Research, Taihe HospitalHubei University of MedicineShiyanChina
| | - Jianguo Liu
- Zunyi Medical UniversityZunyiGuizhouChina
- Special Key Laboratory of Oral Diseases ResearchStomatological Hospital Affiliated to Zunyi Medical UniversityGuizhouChina
| |
Collapse
|
41
|
Gaździcka J, Gołąbek K, Strzelczyk JK, Ostrowska Z. Epigenetic Modifications in Head and Neck Cancer. Biochem Genet 2019; 58:213-244. [PMID: 31712935 PMCID: PMC7113219 DOI: 10.1007/s10528-019-09941-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common human malignancy in the world, with high mortality and poor prognosis for patients. Among the risk factors are tobacco and alcohol intake, human papilloma virus, and also genetic and epigenetic modifications. Many studies show that epigenetic events play an important role in HNSCC development and progression, including DNA methylation, chromatin remodeling, histone posttranslational covalent modifications, and effects of non-coding RNA. Epigenetic modifications may influence silencing of tumor suppressor genes by promoter hypermethylation, regulate transcription by microRNAs and changes in chromatin structure, or induce genome instability through hypomethylation. Moreover, getting to better understand aberrant patterns of methylation may provide biomarkers for early detection and diagnosis, while knowledge about target genes of microRNAs may improve the therapy of HNSCC and extend overall survival. The aim of this review is to present recent studies which demonstrate the role of epigenetic regulation in the development of HNSCC.
Collapse
Affiliation(s)
- Jadwiga Gaździcka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808, Zabrze, Katowice, Poland.
| | - Karolina Gołąbek
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808, Zabrze, Katowice, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808, Zabrze, Katowice, Poland
| | - Zofia Ostrowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Jordana 19 Str., 41-808, Zabrze, Katowice, Poland
| |
Collapse
|
42
|
Potential of Melatonin as Adjuvant Therapy of Oral Cancer in the Era of Epigenomics. Cancers (Basel) 2019; 11:cancers11111712. [PMID: 31684096 PMCID: PMC6895876 DOI: 10.3390/cancers11111712] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/16/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
The wide variety of epigenetic controls available is rapidly expanding the knowledge of molecular biology even overflowing it. At the same time, it can illuminate unsuspected ways of understanding the etiology of cancer. New emerging therapeutic horizons, then, promise to overcome the current antitumor strategies need. The translational utility of this complexity is particularly welcome in oral cancer (OC), in which natural history is alarmingly disappointing due to the invasive and mutilating surgery, the high relapsing rate, the poor quality of life and the reduced survival after diagnosis. Melatonin activates protective receptor-dependent and receptor-independent processes that prevent tissue cancerisation and inhibit progressive tumor malignancy and metastasis. Related evidence has shown that melatonin pleiotropy encompasses gene expression regulation through all the three best-characterized epigenetic mechanisms: DNA methylation, chromatin modification, and non-coding RNA. OC has received less attention than other cancers despite prognosis is usually negative and there are no significant therapy improvements recorded in the past decade. However, a large research effort is being carried out to elucidate how melatonin´s machinery can prevent epigenetic insults that lead to cancer. In the light of recent findings, a comprehensive examination of biochemistry through which melatonin may reverse epigenetic aberrations in OC is an extraordinary opportunity to take a step forward in the clinical management of patients.
Collapse
|
43
|
Gissi DB, Gabusi A, Tarsitano A, Rossi R, Balbi T, Morandi L. Multi-Region Sequence Analysis of a Pregnancy-Related Oral Squamous Cell Carcinoma Exhibiting Low-Level Aggressive Behavior. Int J Surg Pathol 2019; 28:188-195. [PMID: 31544567 DOI: 10.1177/1066896919876058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We analyzed the genetic and epigenetic profiles of an oral squamous cell carcinoma affecting a 41-year-old pregnant female. The patient presented with an oral mass located at the hard and soft palate with bone involvement and lymph node metastases (T4N1M0). She had been treated with multimodal radiotherapy and chemotherapy, and she is currently alive with no evidence of disease 8 years after treatment. DNA methylation and DNA mutation analyses were used to analyze multiple samples from the tumor mass and from the non-neoplastic mucosa to verify tumor heterogeneity. Genetic and epigenetic analyses revealed the presence of one shared TP53 driver mutation with the same DNA methylation profile in each of the 3 areas of the tumor mass; only 2 additional passenger mutations were detected, suggesting a simple clonal homogeneous carcinoma, which usually is associated with low-level aggressive behavior. Additionally, no genetic or epigenetic alteration in the non-neoplastic oral mucosa was detected, demonstrating the absence of field cancerization. The low aggressiveness of the lesion was confirmed by the patient being free of disease at a long-term follow-up examination. These data suggest a different molecular transformation pathway in pregnancy-related oral squamous cell carcinomas, providing new perspectives for further investigation.
Collapse
|
44
|
PD-1 (PDCD1) promoter methylation in Merkel cell carcinoma: prognostic relevance and relationship with clinico-pathological parameters. Mod Pathol 2019; 32:1359-1372. [PMID: 30976104 DOI: 10.1038/s41379-019-0261-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/08/2019] [Accepted: 02/26/2019] [Indexed: 12/23/2022]
Abstract
Merkel cell carcinoma is an aggressive neuroendocrine skin tumor, for which several non-conclusive prognostic factors of adverse clinical behavior have been reported. As promoter methylation of the immune checkpoint receptor CD279/PD-1/PDCD1(mPDCD1) has been shown to be a prognostic factor in different cancers, we investigated its role in Merkel cell carcinoma. mPDCD1was assessed retrospectively in a cohort of 69 Merkel cell carcinoma patients from the University of Bologna, University of Turin and University of Insubria. Kaplan-Meier curves and log-rank tests were calculated for all variables. To assess the influence of mPDCD1, the Cox proportional hazards model and different Royston-Parmar models were evaluated. High PDCD1 methylation (mPDCD1high) was associated with a higher overall mortality at both the univariate analysis (log rank test: χ2 = 5.17, p = 0.023; permutation test: p = 0.023) and the multivariate analysis (HR = 2.111, p = 0.042). The other variables associated with a higher overall mortality at the multivariate analysis were clinical stage III-IV (HR = 2.357, p = 0.008), size > 2 cm (HR = 2.248, p = 0.031) and Merkel cell polyomavirus (HR = 0.397, p = 0.015). Further, mPDCD1high was strongly associated with older age (81 vs 76 years, p = 0.042), absence of immune cells (92.6%, p < 0.001), no expression of PD-L1 by immune cells (70.4%, p = 0.041) and by both immune and tumor cells (70.4%, p = 0.001). mPDCD1 is a valid prognostic parameter in patients affected by Merkel cell carcinoma. In addition, it could provide an estimate of the global PD-1/PD-L1 expression with potentially relevant implications from a therapeutic point of view.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The present review aims to describe the epigenetic alterations observed in oral cancer linked to the exposure to alcohol and/or tobacco. RECENT FINDINGS Recent findings emphasize the importance of epigenetics in oral cancer progression and in how risk factors (as tobacco and alcohol) affect the basal epigenetic profiles. Deeper techniques and detailed approaches allowed the perception that individual CG changes and even subtle changes may represent important epigenetic alterations resulting in expression changes and other carcinogenic consequences. New classes of epigenetic alterations including noncoding RNAs have been gaining attention. SUMMARY Many epigenetic alterations have been described in oral carcinoma progression induced by tobacco and/or alcohol, including: promoter hypermethylation in genes with tumor suppressive activity, global (genome-wide) hypomethylation, change in methylation patterns throughout the genes, alteration in noncoding RNAs, and histones modifications. These changes represent progress in the knowledge of how these risk factors act in a molecular level. There is an urgent need for large independent studies to move these potential makers further and validate them to identify risk assessment, early diagnostic markers, and therapeutic targets, as well as to be the base for prevention and intervention strategies.
Collapse
|
46
|
Low LINC00599 expression is a poor prognostic factor in glioma. Biosci Rep 2019; 39:BSR20190232. [PMID: 30867254 PMCID: PMC6443953 DOI: 10.1042/bsr20190232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022] Open
Abstract
LINC00599 has been suggested to be involved in physiological and pathological processes including carcinogenesis. However, the clinical and prognostic significance of LINC00599 in glioma patients and the effect of LINC00599 on glioma cell migration and invasion remain unknown. In our results, we first observe the expression of LINC00599 in 31 types of human cancers including tumor tissues and corresponding normal tissues at The Cancer Genome Atlas (TCGA) database, and found that LINC00599 expression levels were only reduced in lower grade glioma (LGG) tissues and glioblastoma multiforme (GBM) tissues compared with normal brain tissues. Moreover, we confirmed levels of LINC00599 expression were decreased in glioma tissues and cell lines compared with matched adjacent normal tissues and normal human astrocytes (NHAs), respectively. Meanwhile, we found that glioma tissues with WHO III-IV grade exhibited lower levels of LINC00599 expression than glioma tissues with I-II grade. The survival analysis at TCGA data showed low LINC00599 expression was associated with poor disease-free survival and overall survival in glioma patients. In vitro study suggested up-regulation of LINC00599 depressed glioma cell migration and invasion through regulating epithelial–mesenchymal transition (EMT) process. In conclusion, LINC00599 acts as a tumor-suppressing long non-coding RNA (lncRNA) in glioma.
Collapse
|
47
|
Guan Z, Yu H, Cuk K, Zhang Y, Brenner H. Whole-Blood DNA Methylation Markers in Early Detection of Breast Cancer: A Systematic Literature Review. Cancer Epidemiol Biomarkers Prev 2018; 28:496-505. [DOI: 10.1158/1055-9965.epi-18-0378] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/09/2018] [Accepted: 11/20/2018] [Indexed: 11/16/2022] Open
|
48
|
A Noninvasive Test for MicroRNA Expression in Oral Squamous Cell Carcinoma. Int J Mol Sci 2018; 19:ijms19061789. [PMID: 29914173 PMCID: PMC6032413 DOI: 10.3390/ijms19061789] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 01/10/2023] Open
Abstract
MicroRNAs have recently been proposed as non-invasive biomarkers in Oral Squamous Cell Carcinoma (OSCC). The aim of this study was to analyze the expression of a panel of miRNAs in epithelial cells collected by oral brushing from OSCCs from regenerative areas after OSCC surgical resection and from their respective normal distant mucosa. Oral brushing specimens were collected from 24 healthy donors, 14 OSCC patients with specimens from tumour and normal distant mucosa, and from 13 patients who had OSCC resection, with samples from regenerative areas after OSCC resection and normal distant mucosa. Expression levels of eight targets (miR-21, miR-375, miR-345, miR-181b, miR-146a, miR-649, miR-518b, and miR-191) were evaluated by real-time Polymerase Chain Reaction (PCR). A highly significant between-group difference was found for miR-21 (F = 6.58, p < 0.001), miR-146a (F = 6.974, p < 0.001), and miR-191 (F = 17.07, p < 0.001). The major difference was observed between samples from healthy donors and from OSCC brushing, whereas no significant differences were observed between areas infiltrated by OSCC and their respective normal distant mucosa. Furthermore, altered expression of miR-146a and miR-191 was also observed in regenerative areas after OSCC resection. CONCLUSIONS Oral brushing could be proposed as a noninvasive method to study microRNA expression in oral mucosa in OSCC patients.
Collapse
|
49
|
Li J, Song P, Jiang T, Dai D, Wang H, Sun J, Zhu L, Xu W, Feng L, Shin VY, Morrison H, Wang X, Jin H. Heat Shock Factor 1 Epigenetically Stimulates Glutaminase-1-Dependent mTOR Activation to Promote Colorectal Carcinogenesis. Mol Ther 2018; 26:1828-1839. [PMID: 29730197 DOI: 10.1016/j.ymthe.2018.04.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/01/2018] [Accepted: 04/10/2018] [Indexed: 01/05/2023] Open
Abstract
Heat shock factor 1 (HSF1) generally exhibits its properties under stress conditions. In tumors, HSF1 has a pleiotropic feature in regulating growth, survival, and aggressiveness of cancer cells. In this study, we found HSF1 was increased in colorectal cancer (CRC) and had a positive correlation with shorter disease-free survival (DFS). Knockdown of HSF1 in CRC cells attenuated their growth while inhibiting mTOR activation and glutamine metabolism. HSF1 inhibited the expression of microRNA137 (MIR137), which targeted GLS1 (glutaminase 1), thus stimulating GLS1 protein expression to promote glutaminolysis and mTOR activation. HSF1 bound DNA methyltransferase DNMT3a and recruited it to the promoter of lncRNA MIR137 host gene (MIR137HG), suppressing the generation of primary MIR137. The chemical inhibitor of HSF1 also reduced cell growth, increased apoptosis, and impaired glutamine metabolism in vitro. Moreover, both chemical inhibition and genetic knockout of HSF1 succeeded in increasing MIR137 expression, reducing GLS1 expression, and alleviating colorectal tumorigenesis in azoxymethane (AOM)/dextran sulfate sodium (DSS) mice. In conclusion, HSF1 expression was increased and associated with poor prognosis in CRC. By recruiting DNMT3a to suppress the expression of MIR137 that targets GLS1 mRNA, HSF1 stimulated GLS1-dependent mTOR activation to promote colorectal carcinogenesis. Therefore, targeting HSF1 to attenuate glutaminolysis and mTOR activation could be a promising approach for CRC treatment.
Collapse
Affiliation(s)
- Jiaqiu Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Ping Song
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Tingting Jiang
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Dongjun Dai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hanying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Jie Sun
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Liyuan Zhu
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wenxia Xu
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Vivian Y Shin
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Zhejiang Biotherapy, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| |
Collapse
|