1
|
Wu N, Wu Z, Wang Y, Zhang A, Peng Y, Cheng Y, Lei H, Liu S, Zhao J, Li T, Zhou G. A diagnostic model for non-invasive urothelial cancer early detection based on methylation of urinary tumor DNA. Cancer Cell Int 2025; 25:148. [PMID: 40234942 PMCID: PMC12001437 DOI: 10.1186/s12935-025-03766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Diagnostic methods for urothelial cancer (UC) are often invasive, while urinary cytology, a non-invasive alternative, suffers from limited sensitivity. This study aimed to identify differentially methylated markers in urinary tumor DNA and develop a diagnostic method to enhance the sensitivity of non-invasive UC detection. METHODS Whole-genome bisulfite sequencing and deep methylation sequencing were employed to identify significantly hypermethylated UC-associated genes in clinical samples and public UC datasets. Further screening was conducted using tumor biopsies and urine samples from patients, leading to the selection of three hypermethylated UC markers. A diagnostic model based on these markers was constructed and validated in a cohort (N = 432) comprising patients with UC, other cancers, benign lesions, and non-UC urinary tract diseases. RESULTS Validation in a cohort of 432 subjects demonstrated that the UC diagnostic model, incorporating three hypermethylated markers (VIM, TMEM220, and PPM1N), achieved an overall sensitivity of 94.44% in 108 UC patients. Specificities were 96.34%, 90.76%, and 87.72% in 191 non-neoplastic individuals, 76 patients with benign lesions, and 57 patients with other cancers, respectively, resulting in an overall specificity of 93.52%. Methylation level analysis revealed significantly higher methylation (P < 0.001) for three markers in UC samples compared to non-UC samples. Furthermore, the model exhibited sensitivities of 80% and 88.57% for detecting stage 0a/0is and stage I UC, respectively. CONCLUSIONS The UC diagnostic model demonstrates excellent diagnostic performance, particularly in the early detection of UC. This non-invasive approach, characterized by high sensitivity and specificity, holds significant potential for further clinical evaluation and development as a reliable tool for UC diagnosis using urine samples.
Collapse
Affiliation(s)
- Ningning Wu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Zhen Wu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Yanwen Wang
- Department of Urology, The First Affiliated Hospital of Dali University, 32 Carlsberg Avenue, Dali City, 671000, Yunnan Province, China
| | - Anqi Zhang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Yongfei Peng
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Yan Cheng
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Hongsong Lei
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, 374 Yunnan-Burma Road, Wuhua District, Kunming, 650000, Yunnan Province, China
| | - Siwen Liu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Jie Zhao
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| | - Tianbao Li
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| | - Guangpeng Zhou
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| |
Collapse
|
2
|
Laun SE, Kann L, Braun J, Pierre F, Kim S, Gilbert S, Lunz D, Kalra A, Ma K, Cheng Y, Leggett CL, Zaidi AH, Omstead AN, Korman L, Jobe B, Perpetua L, Greenwald BD, Maddala T, Meltzer SJ. Spatiotemporal Study of a Risk-Stratification Epigenetic-Based Biomarker Assay in Patients With Barrett Esophagus. Am J Gastroenterol 2025:00000434-990000000-01586. [PMID: 39933887 DOI: 10.14309/ajg.0000000000003367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION Barrett esophagus (BE) is the strongest known risk factor for developing esophageal adenocarcinoma (EAC), the second-most lethal cancer in the United States. Esopredict is a novel validated methylation-based biomarker assay that provides precise quantification of neoplastic progression risk in BE patients. Inherit challenges, including tissue heterogeneity, sampling error, interobserver variability, and inconsistent adherence to surveillance biopsy guidelines, may affect the predictive value results of Esopredict obtained at different anatomic locations or different sampling time points. METHODS To investigate the spatiotemporal performance of Esopredict across multiple spatiotemporal sampling points, we profiled 220 biopsies obtained from 58 BE patients, including 11 patients with overlapping spatial and temporal biopsies. We focused on spatial profiling (i.e., multiple biopsies obtained at several anatomic locations during a single endoscopy) and temporal profiling (i.e., biopsies obtained from multiple endoscopies performed at different time points). Each patient had an initial histologic diagnosis of nondysplastic Barrett esophagus, indefinite for dysplasia, or low-grade dysplasia. Final follow-up (endpoint) biopsies showed either high-grade dysplasia or EAC (progressors), or nondysplastic Barrett esophagus, indefinite for dysplasia, or low-grade dysplasia (nonprogressors). Biopsies were analyzed with Esopredict to compute a progression risk score, which quantified the likelihood of future progression to high-grade dysplasia or EAC within 5 years. RESULTS In 52 spatially profiled patients, Esopredict demonstrated a sensitivity of 81% (17/21 progressor patients), based on the highest-scoring biopsy from each patient; sensitivity increased to 100% (12/12) when end point biopsies occurred within 5 years of the index (initial) biopsy. In 28 temporally profiled patients, sensitivity was 100% (8/8 patients), based on the biopsy performed at the time point closest to the end point biopsy. DISCUSSION Esopredict showed high predictive performance in multiple spatiotemporal samples in BE patients. These data further support the use of Esopredict as a robust test to distinguish high-risk BE patients, who may benefit from endoscopic eradication therapy or increased surveillance frequency, from low-risk patients, who may be candidates for less frequent surveillance and noninterventional observation.
Collapse
Affiliation(s)
| | | | | | | | - Suji Kim
- Previse, Baltimore, Maryland, USA
| | | | | | - Andrew Kalra
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ke Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Jefferson Einstein Philadelphia Hospital, Philadelphia, Pennsylvania, USA
| | - Yulan Cheng
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cadman L Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ali H Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Ashten N Omstead
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Louis Korman
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Blair Jobe
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Esophageal Institute, Allegheny Health Network, Pittsburg, Pennsylvania, USA
| | - Lorrie Perpetua
- Research Tissue Biorepository Core Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Bruce D Greenwald
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J Meltzer
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Tang Z, Wu YP, Tan BG, Chen XQ, Guo WW, Wu KS, Zhang XM, Chen TW, Zhou HY. Apparent diffusion coefficient and its standard deviation from diffusion-weighted imaging in preoperative predicting liver invasion by T3-staged resectable gallbladder carcinoma. Clin Radiol 2024; 79:e247-e255. [PMID: 38007337 DOI: 10.1016/j.crad.2023.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/27/2023]
Abstract
AIM To evaluate apparent diffusion coefficient (ADC) and its standard deviation (SDADC) in preoperative predicting liver invasion by T3-staged gallbladder carcinoma (GBC). MATERIALS AND METHODS Forty-one consecutive patients with T3-staged resectable GBC were included and divided into two sets with (n=27) and without (n=14) liver invasion. All patients underwent DWI at b-values of 0, 20, 50, 80, 100, 200, 400, 600, 800, and 1,000 s/mm2 with a 3 T magnetic resonance imaging scanner before surgery. ADC and SDADC of tumour-adjacent and tumour-distant liver tissues were measured on DWI, and were compared by Mann-Whitney U-tests. If there was a significant difference in any derived parameter, the area under the receiver operating characteristic curve (AUC) was used to assess performance of this parameter to predict liver invasion. RESULTS DWI could differentiate between patients with and without liver invasion when b = 0, 1,000 s/mm2 (AUCs of ADC and SDADC were 0.697 and 0.714, respectively). In patients with liver invasion, mean ADC and SDADC of tumour-adjacent liver tissue were lower than of tumour-distant liver tissue when b = 0, 800 s/mm2, and = 0, 1,000 s/mm2 (all p-values <0.05). To differentiate tumour-adjacent from tumour-distant liver tissues in patients with liver invasion, AUCs of ADC were 0.687 (b = 0, 800 s/mm2) and 0.680 (b = 0, 1,000 s/mm2), and AUCs of SDADC were 0.673 (b = 0, 800 s/mm2) and 0.731 (b = 0, 1,000 s/mm2). CONCLUSIONS DWI could have potential value in preoperative predicting liver invasion by T3-staged GBC.
Collapse
Affiliation(s)
- Z Tang
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Department of Radiology, Meishan Hospital of Traditional Chinese Medicine, Meishan, Sichuan, China
| | - Y-P Wu
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China
| | - B-G Tan
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Department of Radiology, Panzhihua Central Hospital, Panzhihua, Sichuan, China
| | - X-Q Chen
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - W-W Guo
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - K-S Wu
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - X-M Zhang
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - T-W Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, China.
| | - H-Y Zhou
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
4
|
Geissler F, Nesic K, Kondrashova O, Dobrovic A, Swisher EM, Scott CL, J. Wakefield M. The role of aberrant DNA methylation in cancer initiation and clinical impacts. Ther Adv Med Oncol 2024; 16:17588359231220511. [PMID: 38293277 PMCID: PMC10826407 DOI: 10.1177/17588359231220511] [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: 08/02/2023] [Accepted: 11/21/2023] [Indexed: 02/01/2024] Open
Abstract
Epigenetic alterations, including aberrant DNA methylation, are now recognized as bone fide hallmarks of cancer, which can contribute to cancer initiation, progression, therapy responses and therapy resistance. Methylation of gene promoters can have a range of impacts on cancer risk, clinical stratification and therapeutic outcomes. We provide several important examples of genes, which can be silenced or activated by promoter methylation and highlight their clinical implications. These include the mismatch DNA repair genes MLH1 and MSH2, homologous recombination DNA repair genes BRCA1 and RAD51C, the TERT oncogene and genes within the P15/P16/RB1/E2F tumour suppressor axis. We also discuss how these methylation changes might occur in the first place - whether in the context of the CpG island methylator phenotype or constitutional DNA methylation. The choice of assay used to measure methylation can have a significant impact on interpretation of methylation states, and some examples where this can influence clinical decision-making are presented. Aberrant DNA methylation patterns in circulating tumour DNA (ctDNA) are also showing great promise in the context of non-invasive cancer detection and monitoring using liquid biopsies; however, caution must be taken in interpreting these results in cases where constitutional methylation may be present. Thus, this review aims to provide researchers and clinicians with a comprehensive summary of this broad, but important subject, illustrating the potentials and pitfalls of assessing aberrant DNA methylation in cancer.
Collapse
Affiliation(s)
- Franziska Geissler
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ksenija Nesic
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Olga Kondrashova
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alexander Dobrovic
- University of Melbourne Department of Surgery, Austin Health, Heidelberg, VIC, Australia
| | | | - Clare L. Scott
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
- Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Matthew J. Wakefield
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
5
|
Tihagam RD, Bhatnagar S. Detection Methods for Epigenetic Mechanisms in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1465:99-103. [PMID: 39586996 DOI: 10.1007/978-3-031-66686-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Epigenetic dysregulation of gene expression is central to breast cancer initiation, progression, and treatment response. These alterations include histone modifications, DNA methylation, and expression of noncoding RNA. Technological advancements have improved our ability to identify histone modifications, DNA methylation patterns and provided critical insights into the epigenetic regulation of gene expression and its role in breast oncogenesis. The epigenetic profiles of healthy and breast cancer tissues revealed several diagnostic and prognostic biomarkers. In this article, we review the methodologies commonly used to study tumor-associated histone modifications and DNA methylation changes in breast cancer, highlighting their principles, applications, and advancements, such as chromatin immunoprecipitation (ChIP), bisulfite conversion of DNA, next-generation sequencing (NGS), and chromatin immunoprecipitation sequencing (ChIP-seq).
Collapse
Affiliation(s)
- Rachisan Djiake Tihagam
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA.
| |
Collapse
|
6
|
Botezatu IV, Kondratova VN, Stroganova AM, Dranko SL, Lichtenstein AV. Aberrant methylation scanning by quantitative DNA melting analysis with hybridization probes as exemplified by liquid biopsy of SEPT9 and HIST1H4F in colorectal cancer. Clin Chim Acta 2023; 551:117591. [PMID: 37832390 DOI: 10.1016/j.cca.2023.117591] [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: 01/10/2023] [Revised: 06/05/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVE The generally accepted method of quantifying hypermethylated DNA by qPCR using methylation-specific primers has the risk of underestimating DNA methylation and requires data normalization. This makes the analysis complicated and less reliable. METHODS The end-point PCR method, called qDMA-HP (for quantitative DNA Melting Analysis with hybridization probes), which excludes the normalization procedure, is multiplexed and quantitative, has been proposed. qDMA-HP is characterized by the following features: (i) asymmetric PCR with methylation-independent primers; (ii) fluorescent dual-labeled, self-quenched probes (commonly known as TaqMan probes) covering several interrogated CpGs; (iii) post-PCR melting analysis of amplicon/probe hybrids; (iv) quantitation of unmethylated and methylated DNA alleles by measuring the areas under the corresponding melt peaks. RESULTS qDMA-HP was tested in liquid biopsy of colorectal cancer by evaluating SEPT9 and HIST1H4F methylations simultaneously in the single-tube reaction. Differences in the methylation levels in healthy donors versus cancer patients were statistically significant (p < 0.0001), AUCROC values were 0.795-0.921 for various marker combinations. CONCLUSIONS This proof-of-concept study shows that qDMA-HP is a simple, normalization-independent, quantitative, multiplex and "closed tube" method easily adapted to clinical settings. It is demonstrated, for the first time, that HIST1H4F is a perspective marker for liquid biopsy of colorectal cancer.
Collapse
Affiliation(s)
- Irina V Botezatu
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, Moscow 115478, Russia
| | - Valentina N Kondratova
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, Moscow 115478, Russia
| | - Anna M Stroganova
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, Moscow 115478, Russia
| | - Svetlana L Dranko
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, Moscow 115478, Russia
| | - Anatoly V Lichtenstein
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, Moscow 115478, Russia.
| |
Collapse
|
7
|
Chen H, Li X, Li F, Li Y, Chen F, Zhang L, Ye F, Gong M, Bu H. Prediction of coexisting invasive carcinoma on ductal carcinoma in situ (DCIS) lesions by mass spectrometry imaging. J Pathol 2023; 261:125-138. [PMID: 37555360 DOI: 10.1002/path.6154] [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: 10/15/2022] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 08/10/2023]
Abstract
Due to limited biopsy samples, ~20% of DCIS lesions confirmed by biopsy are upgraded to invasive ductal carcinoma (IDC) upon surgical resection. Avoiding underestimation of IDC when diagnosing DCIS has become an urgent challenge in an era discouraging overtreatment of DCIS. In this study, the metabolic profiles of 284 fresh frozen breast samples, including tumor tissues and adjacent benign tissues (ABTs) and distant surrounding tissues (DSTs), were analyzed using desorption electrospray ionization-mass spectrometry (DESI-MS) imaging. Metabolomics analysis using DESI-MS data revealed significant differences in metabolite levels, including small-molecule antioxidants, long-chain polyunsaturated fatty acids (PUFAs) and phospholipids between pure DCIS and IDC. However, the metabolic profile in DCIS with invasive carcinoma components clearly shifts to be closer to adjacent IDC components. For instance, DCIS with invasive carcinoma components showed lower levels of antioxidants and higher levels of free fatty acids compared to pure DCIS. Furthermore, the accumulation of long-chain PUFAs and the phosphatidylinositols (PIs) containing PUFA residues may also be associated with the progression of DCIS. These distinctive metabolic characteristics may offer valuable indications for investigating the malignant potential of DCIS. By combining DESI-MS data with machine learning (ML) methods, various breast lesions were discriminated. Importantly, the pure DCIS components were successfully distinguished from the DCIS components in samples with invasion in postoperative specimens by a Lasso prediction model, achieving an AUC value of 0.851. In addition, pixel-level prediction based on DESI-MS data enabled automatic visualization of tissue properties across whole tissue sections. Summarily, DESI-MS imaging on histopathological sections can provide abundant metabolic information about breast lesions. By analyzing the spatial metabolic characteristics in tissue sections, this technology has the potential to facilitate accurate diagnosis and individualized treatment of DCIS by inferring the presence of IDC components surrounding DCIS lesions. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Hong Chen
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Key Laboratory of Transplant Engineering and Immunology of the National Health Commission, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xin Li
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fengling Li
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yijie Li
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Key Laboratory of Transplant Engineering and Immunology of the National Health Commission, West China Hospital, Sichuan University, Chengdu, PR China
| | - Fei Chen
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Lu Zhang
- Image Processing and Parallel Computing Laboratory, School of Computer Science, Southwest Petroleum University, Chengdu, PR China
| | - Feng Ye
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Key Laboratory of Transplant Engineering and Immunology of the National Health Commission, West China Hospital, Sichuan University, Chengdu, PR China
| | - Meng Gong
- Laboratory of Clinical Proteomics and Metabolomics, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Hong Bu
- Department of Pathology and Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, PR China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
8
|
Soltan MA, Alhanshani AA, Shati AA, Alqahtani YA, Alshaya DS, Alharthi J, Altalhi SA, Fayad E, Zaki MSA, Eid RA. Cyclin Dependent Kinase Inhibitor 2A Genetic and Epigenetic Alterations Interfere with Several Immune Components and Predict Poor Clinical Outcome. Biomedicines 2023; 11:2254. [PMID: 37626750 PMCID: PMC10452213 DOI: 10.3390/biomedicines11082254] [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: 07/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Cyclin dependent kinase inhibitor 2A (CDKN2A) is a well-known tumor suppressor gene as it functions as a cell cycle regulator. While several reports correlate the malfunction of CDKN2A with the initiation and progression of several types of human tumors, there is a lack of a comprehensive study that analyzes the potential effect of CDKN2A genetic alterations on the human immune components and the consequences of that effect on tumor progression and patient survival in a pan-cancer model. The first stage of the current study was the analysis of CDKN2A differential expression in tumor tissues and the corresponding normal ones and correlating that with tumor stage, grade, metastasis, and clinical outcome. Next, a detailed profile of CDKN2A genetic alteration under tumor conditions was described and assessed for its effect on the status of different human immune components. CDKN2A was found to be upregulated in cancerous tissues versus normal ones and that predicted the progression of tumor stage, grade, and metastasis in addition to poor prognosis under different forms of tumors. Additionally, CDKN2A experienced different forms of genetic alteration under tumor conditions, a characteristic that influenced the infiltration and the status of CD8, the chemokine CCL4, and the chemokine receptor CCR6. Collectively, the current study demonstrates the potential employment of CDKN2A genetic alteration as a prognostic and immunological biomarker under several types of human cancers.
Collapse
Affiliation(s)
- Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia 41611, Egypt
| | - Ahmad A. Alhanshani
- Department of Child Health, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Ayed A. Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Youssef A. Alqahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Dalal Sulaiman Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Jawaher Alharthi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sarah Awwadh Altalhi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| |
Collapse
|
9
|
Gunawardena K, Sirisena ND, Anandagoda G, Neththikumara N, Dissanayake VHW. Germline variants of uncertain significance, their frequency, and clinico-pathological features in a cohort of Sri Lankan patients with hereditary breast cancer. BMC Res Notes 2023; 16:95. [PMID: 37277882 DOI: 10.1186/s13104-023-06365-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Next-Generation Sequencing (NGS)-based testing in cancer patients has led to increased detection of variants of uncertain significance (VUS). VUS are genetic variants whose impact on protein function is unknown. VUS pose a challenge to clinicians and patients due to uncertainty regarding their cancer predisposition risk. Paucity of data exists on the pattern of VUS in under-represented populations. This study describes the frequency of germline VUS and clinico-pathological features in Sri Lankan hereditary breast cancer patients. METHODS Data of 72 hereditary breast cancer patients who underwent NGS-based testing between January 2015 and December 2021 were maintained prospectively in a database and analyzed retrospectively. Data were subjected to bioinformatics analysis and variants were classified according to international guidelines. RESULTS Germline variants were detected in 33/72(45.8%) patients, comprising 16(48.5%) pathogenic/likely pathogenic variants and 17(51.5%) VUS. Distribution of VUS in breast cancer predisposing genes were :APC:1(5.8%), ATM:2(11.7%), BRCA1:1(5.8%), BRCA2:5(29.4%), BRIP1:1(5.8%), CDKN2A:1(5.8%), CHEK2:2(11.7%), FANC1:1(5.8%), MET:1(5.8%), STK11:1(5.8%), NF2:1(5.8%). Mean age at cancer diagnosis in patients with VUS was 51.2 years. Most common tumour histopathology was ductal carcinoma 11(78.6%). 50% of tumours in patients having VUS in BRCA1/2 genes were hormone receptor negative. 73.3% patients had family history of breast cancer. CONCLUSIONS A significant portion of patients had a germline VUS. Highest frequency was in BRCA2 gene. Majority had family history of breast cancer. This highlights the need to undertake functional genomic studies to determine the biological effects of VUS and identify potentially clinically actionable variants that would be useful for decision-making and patient management.
Collapse
Affiliation(s)
- Kawmadi Gunawardena
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka
| | - Nirmala D Sirisena
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka.
| | - Gayani Anandagoda
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka
| | - Nilaksha Neththikumara
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka
| | - Vajira H W Dissanayake
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo 8, Sri Lanka
| |
Collapse
|
10
|
Zappe K, Kopic A, Scheichel A, Schier AK, Schmidt LE, Borutzki Y, Miedl H, Schreiber M, Mendrina T, Pirker C, Pfeiler G, Hacker S, Haslik W, Pils D, Bileck A, Gerner C, Meier-Menches S, Heffeter P, Cichna-Markl M. Aberrant DNA Methylation, Expression, and Occurrence of Transcript Variants of the ABC Transporter ABCA7 in Breast Cancer. Cells 2023; 12:1462. [PMID: 37296582 PMCID: PMC10252461 DOI: 10.3390/cells12111462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
The ABC transporter ABCA7 has been found to be aberrantly expressed in a variety of cancer types, including breast cancer. We searched for specific epigenetic and genetic alterations and alternative splicing variants of ABCA7 in breast cancer and investigated whether these alterations are associated with ABCA7 expression. By analyzing tumor tissues from breast cancer patients, we found CpGs at the exon 5-intron 5 boundary aberrantly methylated in a molecular subtype-specific manner. The detection of altered DNA methylation in tumor-adjacent tissues suggests epigenetic field cancerization. In breast cancer cell lines, DNA methylation levels of CpGs in promoter-exon 1, intron 1, and at the exon 5-intron 5 boundary were not correlated with ABCA7 mRNA levels. By qPCR involving intron-specific and intron-flanking primers, we identified intron-containing ABCA7 mRNA transcripts. The occurrence of intron-containing transcripts was neither molecular subtype-specific nor directly correlated with DNA methylation at the respective exon-intron boundaries. Treatment of breast cancer cell lines MCF-7, BT-474, SK-BR3, and MDA-MB-231 with doxorubicin or paclitaxel for 72 h resulted in altered ABCA7 intron levels. Shotgun proteomics revealed that an increase in intron-containing transcripts was associated with significant dysregulation of splicing factors linked to alternative splicing.
Collapse
Affiliation(s)
- Katja Zappe
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Antonio Kopic
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Alexandra Scheichel
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Ann-Katrin Schier
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Lukas Emanuel Schmidt
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Yasmin Borutzki
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Heidi Miedl
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Schreiber
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Theresa Mendrina
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Georg Pfeiler
- Division of Gynecology and Gynecological Oncology, Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Hacker
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Werner Haslik
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Pils
- Division of Visceral Surgery, Department of General Surgery and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Samuel Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Margit Cichna-Markl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
11
|
Chen XQ, Tan BG, Xu M, Zhou HY, Ou J, Zhang XM, Yu ZY, Chen TW. Apparent diffusion coefficient derived from diffusion-weighted imaging to differentiate between tumor, tumor-adjacent and tumor-distant tissues in resectable rectal adenocarcinoma. Eur J Radiol 2022; 155:110506. [PMID: 36087424 DOI: 10.1016/j.ejrad.2022.110506] [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: 06/23/2022] [Revised: 08/04/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022]
Abstract
PURPOSE To evaluate feasibility of apparent diffusion coefficient (ADC) at different b-values to differentiate between tumor, tumor-adjacent and tumor-distant tissues in rectal adenocarcinoma (RA). MATERIALS AND METHODS Seventy consecutive patients with RA undergoing preoperative diffusion-weighted imaging were retrospectively enrolled. ADCs of tumor, proximal tumor-adjacent tissue (PTA) and tumor-distant tissue (PTD), and distal tumor-adjacent tissue (DTA) and tumor-distant tissue (DTD) were calculated with b-values of 0 and 800 sec/mm2, 0 and 1000 sec/mm2, 0 and 1500 sec/mm2, and multiple b-values of 0, 50, 100, 800, 1000 and 1500 sec/mm2. Statistical analysis was performed to determine feasibility of ADC to differentiate between pairwise tissues. RESULTS Mean ADC of tumor was lower than those of PTA, PTD, DTA and DTD; and mean ADCs of PTA and DTA were lower than those of PTD and DTD at all b-values, respectively (all P-values < 0.001). ADC cut-offs of 1.089 × 10-3 mm2/sec (b = 0, 1000 sec/mm2) or 1.215 × 10-3 mm2/sec (b = 0, 800 sec/mm2), and 1.142 × 10-3 mm2/sec (b = 0, 1000 sec/mm2) or 0.995 × 10-3 mm2/sec (b = 0, 1500 sec/mm2) achieved excellent performance in differentiating tumor from PTA or PTD, and tumor from DTA or DTD (area under receiver operating characteristic curves [AUCs]: 0.813 or 0.952, and 0.970 or 0.996), respectively. ADC cut-offs of 1.625 × 10-3 mm2/sec (b = 0, 800 sec/mm2), and 1.165 × 10-3 mm2/sec (b = 0, 1500 sec/mm2) could differentiate PTA from PTD, and DTA from DTD (AUCs: 0.709 and 0.673), respectively. CONCLUSION ADC can help differentiate between tumor, tumor-adjacent and tumor-distant tissues in RA.
Collapse
Affiliation(s)
- Xiao-Qian Chen
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China
| | - Bang-Guo Tan
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China; Department of Radiology, Panzhihua Central Hospital, 34# Yikang Street, East District, Panzhihua 617067, Sichuan, China
| | - Min Xu
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China
| | - Hai-Ying Zhou
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China.
| | - Jing Ou
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China
| | - Xiao-Ming Zhang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China
| | - Zi-Yi Yu
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China
| | - Tian-Wu Chen
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1# Maoyuan South Road, Shunqing District, Nanchong 637000, Sichuan, China.
| |
Collapse
|
12
|
Mbatha S, Hull R, Dlamini Z. Exploiting the Molecular Basis of Oesophageal Cancer for Targeted Therapies and Biomarkers for Drug Response: Guiding Clinical Decision-Making. Biomedicines 2022; 10:biomedicines10102359. [PMID: 36289620 PMCID: PMC9598679 DOI: 10.3390/biomedicines10102359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Worldwide, oesophageal cancer is the sixth leading cause of deaths related to cancer and represents a major health concern. Sub-Saharan Africa is one of the regions of the world with the highest incidence and mortality rates for oesophageal cancer and most of the cases of oesophageal cancer in this region are oesophageal squamous cell carcinoma (OSCC). The development and progression of OSCC is characterized by genomic changes which can be utilized as diagnostic or prognostic markers. These include changes in the expression of various genes involved in signaling pathways that regulate pathways that regulate processes that are related to the hallmarks of cancer, changes in the tumor mutational burden, changes in alternate splicing and changes in the expression of non-coding RNAs such as miRNA. These genomic changes give rise to characteristic profiles of altered proteins, transcriptomes, spliceosomes and genomes which can be used in clinical applications to monitor specific disease related parameters. Some of these profiles are characteristic of more aggressive forms of cancer or are indicative of treatment resistance or tumors that will be difficult to treat or require more specialized specific treatments. In Sub-Saharan region of Africa there is a high incidence of viral infections such as HPV and HIV, which are both risk factors for OSCC. The genomic changes that occur due to these infections can serve as diagnostic markers for OSCC related to viral infection. Clinically this is an important distinction as it influences treatment as well as disease progression and treatment monitoring practices. This underlines the importance of the characterization of the molecular landscape of OSCC in order to provide the best treatment, care, diagnosis and screening options for the management of OSCC.
Collapse
Affiliation(s)
- Sikhumbuzo Mbatha
- SAMRC Precision Oncology Research Unit (PORU), SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
- Department of Surgery, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
- Correspondence: (S.M.); (Z.D.)
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
- Correspondence: (S.M.); (Z.D.)
| |
Collapse
|
13
|
Pallotti F, Senofonte G, Konstantinidou F, Di Chiano S, Faja F, Rizzo F, Cargnelutti F, Krausz C, Paoli D, Lenzi A, Stuppia L, Gatta V, Lombardo F. Epigenetic Effects of Gender-Affirming Hormone Treatment: A Pilot Study of the ESR2 Promoter’s Methylation in AFAB People. Biomedicines 2022; 10:biomedicines10020459. [PMID: 35203670 PMCID: PMC8962414 DOI: 10.3390/biomedicines10020459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
Virilization of gender-incongruent subjects to whom were assigned the female gender at birth (AFAB) is achieved through testosterone administration. Inter-individual differences in the timing and acquisition of phenotypic characteristics, even if the same hormone preparations and regimens are used, are frequently observed. Polymorphisms of sex hormone receptors and methylation of their gene promoters, as well of several imprinted genes as H19, may underlie the differential response to treatment. Thus, the aim of this study was to examine the possible relationship between the CpG methylation profile of the estrogen receptor 2 gene (ESR2) and H19 promoters and their influence on phenotype modifications in a cohort of AFAB people at baseline (T0) and after 6 mo (T6) and 12 mo (T12) of testosterone therapy (testosterone enanthate, 250 mg i.m. every 28 d). A total of 13 AFAB subjects (mean age 29.3 ± 12.6) were recruited. The percentage of methylation of the ESR2 promoter significantly increased at T6 (adj. p = 0.001) and T12 (adj. p = 0.05), while no difference was detected for H19 (p = 0.237). Methylation levels were not associated with androgen receptor (AR)/estrogen receptor beta (ERβ) polymorphisms nor hormone levels at baseline and after six months of treatment. On the other hand, total testosterone level and patient age resulted in being significantly associated with ESR2 methylation after twelve months of treatment. Finally, the difference in ESR2 promoter methylation between T6 and baseline was significantly associated with the number of CA repeats of the ERβ receptor, adjusted vs. all considered variables (R2 = 0.62, adj. R2 = 0.35). No associations were found with CAG repeats of the AR, age, and estradiol and testosterone levels. Despite the small sample size, we can hypothesize that treatment with exogenous testosterone can modify the ESR2 methylation pattern. Our data also indicated that epigenetic changes may be regulated, suggesting that the modulation of estrogen signaling is relevant shortly after the beginning of the treatment up to T6, with no further significant modification at T12. Furthermore, estrogen receptor methylation appears to be associated with the age of the subjects and exogenous testosterone administration, representing a marker of androgenic treatment. Nonetheless, it will be necessary to increase the number of subjects to evaluate how epigenetic regulation might play a relevant role in the modulation of phenotypical changes after testosterone treatment.
Collapse
Affiliation(s)
- Francesco Pallotti
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Giulia Senofonte
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Fani Konstantinidou
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (L.S.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Silvia Di Chiano
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Fabiana Faja
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Flavio Rizzo
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Francesco Cargnelutti
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy;
| | - Donatella Paoli
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Andrea Lenzi
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (L.S.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (L.S.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence:
| | - Francesco Lombardo
- Laboratory of Seminology—Sperm Bank “Loredana Gandini”, Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (F.P.); (G.S.); (S.D.C.); (F.F.); (F.R.); (F.C.); (D.P.); (A.L.); (F.L.)
| |
Collapse
|
14
|
Mendaza S, Guerrero-Setas D, Monreal-Santesteban I, Ulazia-Garmendia A, Cordoba Iturriagagoitia A, De la Cruz S, Martín-Sánchez E. A DNA Methylation-Based Gene Signature Can Predict Triple-Negative Breast Cancer Diagnosis. Biomedicines 2021; 9:1394. [PMID: 34680511 PMCID: PMC8533184 DOI: 10.3390/biomedicines9101394] [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: 07/19/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer (BC) subtype and lacks targeted treatment. It is diagnosed by the absence of immunohistochemical expression of several biomarkers, but this method still displays some interlaboratory variability. DNA methylome aberrations are common in BC, thereby methylation profiling could provide the identification of accurate TNBC diagnosis biomarkers. Here, we generated a signature of differentially methylated probes with class prediction ability between 5 non-neoplastic breast and 7 TNBC tissues (error rate = 0.083). The robustness of this signature was corroborated in larger cohorts of additional 58 non-neoplastic breast, 93 TNBC, and 150 BC samples from the Gene Expression Omnibus repository, where it yielded an error rate of 0.006. Furthermore, we validated by pyrosequencing the hypomethylation of three out of 34 selected probes (FLJ43663, PBX Homeobox 1 (PBX1), and RAS P21 protein activator 3 (RASA3) in 51 TNBC, even at early stages of the disease. Finally, we found significantly lower methylation levels of FLJ43663 in cell free-DNA from the plasma of six TNBC patients than in 15 healthy donors. In conclusion, we report a novel DNA methylation signature with potential predictive value for TNBC diagnosis.
Collapse
Affiliation(s)
- Saioa Mendaza
- Molecular Pathology of Cancer Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (D.G.-S.); (I.M.-S.); (A.U.-G.)
| | - David Guerrero-Setas
- Molecular Pathology of Cancer Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (D.G.-S.); (I.M.-S.); (A.U.-G.)
- Department of Pathology, Complejo Hospitalario de Navarra (CHN), Irunlarrea 3, 31008 Pamplona, Spain;
| | - Iñaki Monreal-Santesteban
- Molecular Pathology of Cancer Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (D.G.-S.); (I.M.-S.); (A.U.-G.)
| | - Ane Ulazia-Garmendia
- Molecular Pathology of Cancer Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (D.G.-S.); (I.M.-S.); (A.U.-G.)
| | | | - Susana De la Cruz
- Department of Medical Oncology, Complejo Hospitalario de Navarra (CHN), Irunlarrea 3, 31008 Pamplona, Spain;
| | - Esperanza Martín-Sánchez
- Molecular Pathology of Cancer Group, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (D.G.-S.); (I.M.-S.); (A.U.-G.)
| |
Collapse
|
15
|
Xu J, Li N, Deng W, Luo S. Discovering the mechanism and involvement of the methylation of cyclin-dependent kinase inhibitor 2A (CDKN2A) gene and its special locus region in gastric cancer. Bioengineered 2021; 12:1286-1298. [PMID: 33896386 PMCID: PMC8806355 DOI: 10.1080/21655979.2021.1887646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cyclin-dependent kinase inhibitor 2A (CDKN2A) gene methylation has been paramount in the development of malignant masses. The purpose of the conducted research was to evaluate the mechanism and involvement of methylation in regards to the CDKN2A gene and the specific locus region in gastric cancer (GC) with comprehensive statistical analysis utilizing statistics acquired from The Cancer Genome Atlas (TCGA) database. Gene Set Enrichment Analysis (GSEA) revealed that the level of CDKN2A gene methylation and its locus in GC tissues was increased compared to para-cancerous tissues. In multivariate analysis, low methylation of CDKN2A gene, cg03079681, cg04026675, cg07562918, and cg13601799 locus were independently linked to better OS. In addition, the methylation of CDKN2A gene, cg00718440, cg03079681, cg04026675, cg07562918, cg10848754, cg14069088 and cg14430974 locus were negative correlated with CDKN2A gene expression. Meanwhile, the methylation of cg12840719 locus was positively correlated with CDKN2A gene expression. GSEA showed that hallmark_kras_signaling_dn, hallmark_myogenesis, and hallmark_epithelial_mesenchymal_transition pathways were enriched in the CDKN2A gene hypermethylation phenotype. Taken together, the low methylation of CDKN2A gene, cg03079681, cg04026675, cg07562918, and cg13601799 locus indicated a better prognosis in GC. The methylation levels of cg14069088 were most negatively correlated with CDKN2A gene expression. Hallmark_kras_signaling_dn, Hallmark_myogenesis, and hallmark_epithelial_mesenchymal_transition pathways might be important in the regulation of CDKN2A gene hypermethylation.
Collapse
Affiliation(s)
- Jiye Xu
- Department of Oncology, Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.,Department of Medical Oncology, Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Ning Li
- Department of Oncology, Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenying Deng
- Department of Oncology, Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Suxia Luo
- Department of Oncology, Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
He Z, Chen Z, Tan M, Elingarami S, Liu Y, Li T, Deng Y, He N, Li S, Fu J, Li W. A review on methods for diagnosis of breast cancer cells and tissues. Cell Prolif 2020; 53:e12822. [PMID: 32530560 PMCID: PMC7377933 DOI: 10.1111/cpr.12822] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer has seriously been threatening physical and mental health of women in the world, and its morbidity and mortality also show clearly upward trend in China over time. Through inquiry, we find that survival rate of patients with early‐stage breast cancer is significantly higher than those with middle‐ and late‐stage breast cancer, hence, it is essential to conduct research to quickly diagnose breast cancer. Until now, many methods for diagnosing breast cancer have been developed, mainly based on imaging and molecular biotechnology examination. These methods have great contributions in screening and confirmation of breast cancer. In this review article, we introduce and elaborate the advances of these methods, and then conclude some gold standard diagnostic methods for certain breast cancer patients. We lastly discuss how to choose the most suitable diagnostic methods for breast cancer patients. In general, this article not only summarizes application and development of these diagnostic methods, but also provides the guidance for researchers who work on diagnosis of breast cancer.
Collapse
Affiliation(s)
- Ziyu He
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Zhu Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China.,State Key Laboratory of Bioelectronics, School of Biological and Medical Engineering, Southeast University, Nanjing, China
| | - Miduo Tan
- Surgery Department of Galactophore, Central Hospital of Zhuzhou City, Zhuzhou, China
| | - Sauli Elingarami
- School of Life Sciences and Bioengineering (LiSBE), The Nelson Mandela African Institution of Science and Technology (NM-AIST), Arusha, Tanzania
| | - Yuan Liu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China.,State Key Laboratory of Bioelectronics, School of Biological and Medical Engineering, Southeast University, Nanjing, China
| | - Taotao Li
- Hunan Provincial Key Lab of Dark Tea and Jin-hua, School of Materials and Chemical Engineering, Hunan City University, Yiyang, China
| | - Yan Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Nongyue He
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China.,State Key Laboratory of Bioelectronics, School of Biological and Medical Engineering, Southeast University, Nanjing, China
| | - Song Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| | - Juan Fu
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, China
| |
Collapse
|
17
|
Delmonico L, Silva Magalhães Costa MA, Gomes RJ, De Oliveira Vieira P, Da Silva ABP, Fournier MV, Scherrer LR, De Azevedo CM, Ornellas MHF, Alves G. Methylation profiling in promoter sequences of ATM and CDKN2A ( p14ARF/p16INK4a ) genes in blood and cfDNA from women with impalpable breast lesions. Oncol Lett 2020; 19:3003-3010. [PMID: 32218857 DOI: 10.3892/ol.2020.11382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/06/2019] [Indexed: 12/22/2022] Open
Abstract
The objective of the present study was to evaluate the epigenetic changes occurring in early stages of breast cancer. The present study investigated the methylation profile of the ATM, p14ARF and p16INK4a promoters in total blood and plasma cell-free DNA (cfDNA) from women with impalpable breast lesions compared with in total blood of a control cohort of women without breast lesions. The samples were evaluated using the methylation-specific PCR method. The Fisher's exact test was used to evaluate statistical significance between the methylation and clinical variables. A total of 111 women were evaluated, including 56 women with impalpable breast cancer (39/56 also had paired plasma cfDNA) and 55 women in the control cohort (55 blood DNA). For blood DNA from women with malignant impalpable breast lesions, p16INK4a exhibited the greatest percentage of methylation (48%), followed by ATM (37.5%) and p14ARF (27%) promoters, regardless of age variation. For plasma cfDNA, the methylation rates for ATM, p14ARF and p16INK4a were 26, 26 and 10%, respectively. The methylation rates for the blood DNA of controls were the lowest for ATM (9%), p14ARF (7%) and p16INK4a (7%). The women with impalpable breast lesions (benign and malignant lesions) exhibited the highest methylation rate, regardless of age, compared with the paired plasma cfDNA and controls. This epigenetic change was statistically significant for the promoters of ATM (P=0.009) and p16INK4a (P=0.001) (impalpable breast lesions vs. control). The present study demonstrated that epigenetic changes occurring in the ATM and CDKN2A genes detectable in liquid biopsy were associated with the development of impalpable breast lesions.
Collapse
Affiliation(s)
- Lucas Delmonico
- LaRBio-Radiation Laboratory in Biology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil
| | | | - Romario José Gomes
- Circulating Biomarkers Laboratory, Department of General Pathology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Pâmella De Oliveira Vieira
- Circulating Biomarkers Laboratory, Department of General Pathology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Ana Beatriz Passos Da Silva
- Circulating Biomarkers Laboratory, Department of General Pathology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | | | | | | | - Maria Helena Faria Ornellas
- Circulating Biomarkers Laboratory, Department of General Pathology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil.,Postgraduate Program in Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Gilda Alves
- Circulating Biomarkers Laboratory, Department of General Pathology, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil.,Postgraduate Program in Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
18
|
Mendaza S, Ulazia-Garmendia A, Monreal-Santesteban I, Córdoba A, de Azúa YR, Aguiar B, Beloqui R, Armendáriz P, Arriola M, Martín-Sánchez E, Guerrero-Setas D. ADAM12 is A Potential Therapeutic Target Regulated by Hypomethylation in Triple-Negative Breast Cancer. Int J Mol Sci 2020; 21:E903. [PMID: 32019179 PMCID: PMC7036924 DOI: 10.3390/ijms21030903] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 02/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and currently lacks any effective targeted therapy. Since epigenetic alterations are a common event in TNBC, DNA methylation profiling can be useful for identifying potential biomarkers and therapeutic targets. Here, genome-wide DNA methylation from eight TNBC and six non-neoplastic tissues was analysed using Illumina Human Methylation 450K BeadChip. Results were validated by pyrosequencing in an independent cohort of 50 TNBC and 24 non-neoplastic samples, where protein expression was also assessed by immunohistochemistry. The functional role of disintegrin and metalloproteinase domain-containing protein 12(ADAM12) in TNBC cell proliferation, migration and drug response was analysed by gene expression silencing with short hairpin RNA. Three genes (Von Willenbrand factor C and Epidermal Growth Factor domain-containing protein (VWCE), tetraspanin-9 (TSPAN9) and ADAM12) were found to be exclusively hypomethylated in TNBC. Furthermore, ADAM12 hypomethylation was associated with a worse outcome in TNBC tissues and was also found in adjacent-to-tumour tissue and, preliminarily, in plasma from TNBC patients. In addition, ADAM12 silencing decreased TNBC cell proliferation and migration and improved doxorubicin sensitivity in TNBC cells. Our results indicate that ADAM12 is a potential therapeutic target and its hypomethylation could be a poor outcome biomarker in TNBC.
Collapse
Affiliation(s)
- Saioa Mendaza
- Molecular Pathology of Cancer Group, Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (S.M.); (A.U.-G.); (D.G.-S.)
| | - Ane Ulazia-Garmendia
- Molecular Pathology of Cancer Group, Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (S.M.); (A.U.-G.); (D.G.-S.)
| | - Iñaki Monreal-Santesteban
- Molecular Pathology of Cancer Group, Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (S.M.); (A.U.-G.); (D.G.-S.)
| | - Alicia Córdoba
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| | - Yerani Ruiz de Azúa
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| | - Begoña Aguiar
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| | - Raquel Beloqui
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| | - Pedro Armendáriz
- Department of Surgery, ComplejoHospitalario de Navarra, (CHN), Irunlarrea 3, 31008, Pamplona, Spain;
| | - Marta Arriola
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| | - Esperanza Martín-Sánchez
- Molecular Pathology of Cancer Group, Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (S.M.); (A.U.-G.); (D.G.-S.)
| | - David Guerrero-Setas
- Molecular Pathology of Cancer Group, Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain; (S.M.); (A.U.-G.); (D.G.-S.)
- Department of Pathology, ComplejoHospitalario de Navarra (CHN), Irunlarrea 3, 31008, Pamplona, Spain; (A.C.); (Y.R.d.A.); (B.A.); (R.B.); (M.A.)
| |
Collapse
|
19
|
Khan R, Aftab A, Tabassum S, Hussain HMJ, Hameed A, Mahmood H, Munir F, Bukhari I. Identification of CDKN2A variants in breast cancer patients in Pakistan. Genes Genet Syst 2019; 94:117-122. [PMID: 31231091 DOI: 10.1266/ggs.18-00053] [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: 11/23/2022] Open
Abstract
The role of cyclin-dependent kinase inhibitor 2A gene (CDKN2A) variants in breast cancer is not well understood, here we investigated their possible effects on breast cancer in Pakistani women attending the NORI Hospital, Islamabad. Direct DNA sequencing of CDKN2A identified an already known polymorphism in the 3' UTR, c.*29G>C (rs11515), in 5.88% patients and two novel variants. One, a deep intronic substitution (c.458-554T>G) in 1.96% patients, is also detected as a compound heterozygous form along with c.*29G>C in 1.96% patients (c.[458-554T>G; *29G>C]). The other is a novel deletion (c.458-82delG) occurring as a compound variant with two other identified variants c.[458-554T>G; 458-82delG; *29G>C] in 1.96% patients. In silico pathogenicity prediction analyses did not predict pathogenic effects on breast cancer for these individual variants. We conclude that variations in CDKN2A are not the major genetic cause of breast cancer in the enrolled Pakistani patients.
Collapse
Affiliation(s)
- Ranjha Khan
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, University of Science and Technology of China
| | - Ayesha Aftab
- Department of Biological Sciences, International Islamic University
| | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University
| | - Hafiz Muhammad Jafar Hussain
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, University of Science and Technology of China
| | - Abdul Hameed
- Institute of Biomedical and Genetic Engineering (IBGE)
| | - Humera Mahmood
- Department of Oncology, Nuclear Medicine Oncology & Radiotherapy Institute (NORI) Hospital
| | - Faiza Munir
- Department of Biological Sciences, International Islamic University
| | - Ihtisham Bukhari
- Translational Research Institute, Henan Provincial People's Hospital, School of Medicine, Henan University
| |
Collapse
|
20
|
Wang S, Jeong HH, Sohn KA. ClearF: a supervised feature scoring method to find biomarkers using class-wise embedding and reconstruction. BMC Med Genomics 2019; 12:95. [PMID: 31296201 PMCID: PMC6624178 DOI: 10.1186/s12920-019-0512-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Feature selection or scoring methods for the detection of biomarkers are essential in bioinformatics. Various feature selection methods have been developed for the detection of biomarkers, and several studies have employed information-theoretic approaches. However, most of these methods generally require a long processing time. In addition, information-theoretic methods discretize continuous features, which is a drawback that can lead to the loss of information. RESULTS In this paper, a novel supervised feature scoring method named ClearF is proposed. The proposed method is suitable for continuous-valued data, which is similar to the principle of feature selection using mutual information, with the added advantage of a reduced computation time. The proposed score calculation is motivated by the association between the reconstruction error and the information-theoretic measurement. Our method is based on class-wise low-dimensional embedding and the resulting reconstruction error. Given multi-class datasets such as a case-control study dataset, low-dimensional embedding is first applied to each class to obtain a compressed representation of the class, and also for the entire dataset. Reconstruction is then performed to calculate the error of each feature and the final score for each feature is defined in terms of the reconstruction errors. The correlation between the information theoretic measurement and the proposed method is demonstrated using a simulation. For performance validation, we compared the classification performance of the proposed method with those of various algorithms on benchmark datasets. CONCLUSIONS The proposed method showed higher accuracy and lower execution time than the other established methods. Moreover, an experiment was conducted on the TCGA breast cancer dataset, and it was confirmed that the genes with the highest scores were highly associated with subtypes of breast cancer.
Collapse
Affiliation(s)
- Sehee Wang
- Department of Computer Engineering, Ajou University, Suwon, 16499 South Korea
| | - Hyun-Hwan Jeong
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030 USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Kyung-Ah Sohn
- Department of Computer Engineering, Ajou University, Suwon, 16499 South Korea
| |
Collapse
|
21
|
Zhu X, Yang H, Lang J, Zhang Y. Ras association domain family protein 1a hypermethylation and PD-L1 expression in ovarian cancer: A retrospective study of 112 cases. Eur J Obstet Gynecol Reprod Biol 2019; 240:103-108. [PMID: 31242460 DOI: 10.1016/j.ejogrb.2019.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study assessed the interrelationships between Ras association domain family protein 1a (RASSF1A) gene hypermethylation, PD-L1 protein expression, and the clinicopathological characteristics of 112 ovarian cancer (OC) samples. METHODS Formalin-fixed paraffin-embedded OC tissue samples from surgical resection were assessed. Bisulfite pyrosequencing and immunohistochemistry were applied to detect RASSF1A gene methylation and PD-L1 protein expression in tumor cells, respectively. RASSF1A gene methylation and PD-L1 protein expression levels were analyzed against clinicopathological features and prognosis through standard statistical methods. RESULTS Of the 112 OC samples, 49.1% (55/112) exhibited RASSF1A gene hypermethylation. The frequency of RASSF1A hypermethylation was significantly higher in nonserous subtype (73.0%), early stage (66.7%), and nonrecurrent OC (62.9%, p < 0.05). Among all samples, 61.6% (69/112) were positive for PD-L1 protein expression in tumor cells. No significant differences in PD-L1 expression were identified for age, menstrual status, histological type, tumor location, grade, stage, lymph node metastasis, or prognosis (p > 0.05). RASSF1A methylation and PD-L1 expression were not correlated (p > 0.05). CONCLUSIONS This was the first study linking RASSF1A hypermethylation variability to PD-L1 expression and clinicopathological characteristics of OC. Epigenetic alteration of RASSF1A was closely associated with nonserous subtype, early stage, and nonrecurrent OC, indicating that RASSF1A hypermethylation may play a role in early detection of OC. Expression of PD-L1 had no relationship with the studied clinicopathological characteristics or RASSF1A hypermethylation in the 112 OC samples.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
22
|
Li J, Rong MH, Dang YW, He RQ, Lin P, Yang H, Li XJ, Xiong DD, Zhang LJ, Qin H, Feng CX, Chen XY, Zhong JC, Ma J, Chen G. Differentially expressed gene profile and relevant pathways of the traditional Chinese medicine cinobufotalin on MCF‑7 breast cancer cells. Mol Med Rep 2019; 19:4256-4270. [PMID: 30896874 PMCID: PMC6471831 DOI: 10.3892/mmr.2019.10062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023] Open
Abstract
Cinobufotalin is a chemical compound extracted from the skin of dried bufo toads that may have curative potential for certain malignancies through different mechanisms; however, these mechanisms remain unexplored in breast cancer. The aim of the present study was to investigate the antitumor mechanism of cinobufotalin in breast cancer by using microarray data and in silico analysis. The microarray data set GSE85871, in which cinobufotalin exerted influences on the MCF‑7 breast cancer cells, was acquired from the Gene Expression Omnibus database, and the differentially expressed genes (DEGs) were analyzed. Subsequently, protein interaction analysis was conducted, which clarified the clinical significance of core genes, and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were used to analyze cinobufotalin‑related pathways. The Connectivity Map (CMAP) database was used to select existing compounds that exhibited curative properties similar to those of cinobufotalin. A total of 1,237 DEGs were identified from breast cancer cells that were treated with cinobufotalin. Two core genes, SRC proto‑oncogene non‑receptor tyrosine kinase and cyclin‑dependent kinase inhibitor 2A, were identified as serving a vital role in the onset and development of breast cancer, and their expression levels were markedly reduced following cinobufotalin treatment as detected by the microarray of GSE85871. It also was revealed that the 'neuroactive ligand‑receptor interaction' and 'calcium signaling' pathways may be crucial for cinobufotalin to perform its functions in breast cancer. Conducting a matching search in CMAP, miconazole and cinobufotalin were indicated to possessed similar molecular mechanisms. In conclusion, cinobufotalin may serve as an effective compound for the treatment of a subtype of breast cancer that is triple positive for the presence of estrogen, progesterone and human epidermal growth factor receptor‑2 receptors, and its mechanism may be related to different pathways. In addition, cinobufotalin is likely to exert its antitumor influences in a similar way as miconazole in MCF‑7 cells.
Collapse
Affiliation(s)
- Jie Li
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530023, P.R. China
| | - Min-Hua Rong
- Research Department, The Affiliated Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Peng Lin
- Ultrasonics Division of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hong Yang
- Ultrasonics Division of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Jiao Li
- PET‑CT, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Dan-Dan Xiong
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Li-Jie Zhang
- Ultrasonics Division of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hui Qin
- Ultrasonics Division of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Cai-Xia Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Yi Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jin-Cai Zhong
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jie Ma
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
23
|
Panis C, Corrêa S, Binato R, Abdelhay E. The Role of Proteomics in Cancer Research. ONCOGENOMICS 2019:31-55. [DOI: 10.1016/b978-0-12-811785-9.00003-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Ye X, Mo M, Xu S, Yang Q, Wu M, Zhang J, Chen B, Li J, Zhong Y, Huang Q, Cai C. The hypermethylation of p16 gene exon 1 and exon 2: potential biomarkers for colorectal cancer and are associated with cancer pathological staging. BMC Cancer 2018; 18:1023. [PMID: 30348132 PMCID: PMC6198490 DOI: 10.1186/s12885-018-4921-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
Abstract
Background Tumor suppressor gene p16 promoter hypermethylation has been widely studied in colorectal cancer (CRC), yet its clinicopathological significance remains controversial. The methylation alterations of other regions within p16 gene are still rarely researched. The present study aimed to explore the methylation changes of p16 gene body in CRC and to find whether they were associated with clinicopathological staging of CRC. Methods Paired colorectal cancer tissues and corresponding adjacent normal tissues from 30 CRC patients were collected. The methylation levels of two CpG islands within p16 gene body, exon 1 and exon 2, were accurately assessed simultaneously by a LC-MS/MS method. The p16 protein expressions were assessed by immunohistochemistry assay. Statistical analyses were carried out using SPSS 17.0 software. Heat-map analysis was carried out by HemI 1.0 software. Results In the present study, CRC tissues showed more highly methylated than adjacent normal tissues at both CpG islands of p16 gene. And exon 2 hypermethylation was higher and more frequent than exon 1. The ROC curve analysis showed that the simultaneous use of both indicators had excellent sensitivity and specificity for distinguishing CRC tissues and adjacent normal tissues. Following, the methylation level of p16 exon 1/2 was negatively related to p16 protein expression. Further correlation analysis revealed that p16 exon 1 hypermethylation was associated with N/Dukes staging (p = 0.033), and p16 exon 2 hypermethylaiton was associated with T staging (p = 0.035). Conclusions The p16 gene body was remarkably hyper-methylated in CRC tissues and associated with p16 protein expression and cancer clinicopathological staging. The combination of p16 exon 1 and exon 2 could better reflect the overall methylation status of p16 gene body and provide potential biomarkers of CRC.
Collapse
Affiliation(s)
- Xiaoxia Ye
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Mingming Mo
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Simin Xu
- Institute of Neurology, Affiliated Hospital, Guangdong Medical University, No. 2 Wenming Dong Road, Zhanjiang, Guangdong, 524023, People's Republic of China
| | - Qingjin Yang
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Minhua Wu
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Junjie Zhang
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Bin Chen
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Jian Li
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Yu Zhong
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Qionglin Huang
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Chun Cai
- Analysis Center, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China.
| |
Collapse
|
25
|
Aftab A, Shahzad S, Hussain HMJ, Khan R, Irum S, Tabassum S. CDKN2A/P16INK4A variants association with breast cancer and their in-silico analysis. Breast Cancer 2018; 26:11-28. [DOI: 10.1007/s12282-018-0894-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
|
26
|
Davalos V, Martinez-Cardus A, Esteller M. The Epigenomic Revolution in Breast Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2163-2174. [DOI: 10.1016/j.ajpath.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 02/09/2023]
|
27
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|