1
|
Meevassana J, Vongsuly CW, Nakbua T, Kamolratanakul S, Thitiwanichpiwong P, Bin-Alee F, Keelawat S, Kitkumthorn N. Selected Alu methylation levels in the gastric carcinogenesis cascade. PeerJ 2025; 13:e19485. [PMID: 40416611 PMCID: PMC12101442 DOI: 10.7717/peerj.19485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/27/2025] [Indexed: 05/27/2025] Open
Abstract
Background Genome-wide hypomethylation, a common epigenetic change that occurs during cancer development, primarily affects repetitive elements, such as Alu repeats. Consequently, Alu repeats can be used as a surrogate marker of genomic hypomethylation. Methods In this study, we aimed to investigate the correlation between Alu methylation levels and the multistage course of gastric carcinogenesis. Results We found that the Alu methylation levels in gastric cancer tissue decreased compared with those in normal gastric tissue, with the change in methylation levels and pattern being most significant between chronic gastritis and intestinal metaplasia. Moreover, Alu methylation levels were not associated with Helicobacter pylori or Epstein-Barr virus infection. Conclusions Finally, our sensitivity and specificity analyses suggested that Alu methylation level can be used to distinguish gastric cancer tissue from normal tissue. Thus, Alu methylation level shows promise as biomarker for gastric cancer diagnosis.
Collapse
Affiliation(s)
- Jiraroch Meevassana
- Center of Excellence in Burn and Wound Care, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chawisa Wanda Vongsuly
- Center of Excellence in Burn and Wound Care, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanchanok Nakbua
- Center of Excellence in Burn and Wound Care, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Fardeela Bin-Alee
- Faculty of Medicine, Princess of Naradhiwas University, Narathiwat, Thailand
| | - Somboon Keelawat
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Wang Y, Liu H, Zhang M, Xu J, Zheng L, Liu P, Chen J, Liu H, Chen C. Epigenetic reprogramming in gastrointestinal cancer: biology and translational perspectives. MedComm (Beijing) 2024; 5:e670. [PMID: 39184862 PMCID: PMC11344282 DOI: 10.1002/mco2.670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/27/2024] Open
Abstract
Gastrointestinal tumors, the second leading cause of human mortality, are characterized by their association with inflammation. Currently, progress in the early diagnosis and effective treatment of gastrointestinal tumors is limited. Recent whole-genome analyses have underscored their profound heterogeneity and extensive genetic and epigenetic reprogramming. Epigenetic reprogramming pertains to dynamic and hereditable alterations in epigenetic patterns, devoid of concurrent modifications in the underlying DNA sequence. Common epigenetic modifications encompass DNA methylation, histone modifications, noncoding RNA, RNA modifications, and chromatin remodeling. These modifications possess the potential to invoke or suppress a multitude of genes associated with cancer, thereby governing the establishment of chromatin configurations characterized by diverse levels of accessibility. This intricate interplay assumes a pivotal and indispensable role in governing the commencement and advancement of gastrointestinal cancer. This article focuses on the impact of epigenetic reprogramming in the initiation and progression of gastric cancer, esophageal cancer, and colorectal cancer, as well as other uncommon gastrointestinal tumors. We elucidate the epigenetic landscape of gastrointestinal tumors, encompassing DNA methylation, histone modifications, chromatin remodeling, and their interrelationships. Besides, this review summarizes the potential diagnostic, therapeutic, and prognostic targets in epigenetic reprogramming, with the aim of assisting clinical treatment strategies.
Collapse
Affiliation(s)
- Yingjie Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Hongyu Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Mengsha Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jing Xu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Liuxian Zheng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Pengpeng Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jingyao Chen
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Hongyu Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Chong Chen
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
3
|
Babaeenezhad E, Rashidipour M, Jangravi Z, Moradi Sarabi M, Shahriary A. Cytotoxic and epigenetic effects of berberine-loaded chitosan/pectin nanoparticles on AGS gastric cancer cells: Role of the miR-185-5p/KLF7 axis, DNMTs, and global DNA methylation. Int J Biol Macromol 2024; 260:129618. [PMID: 38253156 DOI: 10.1016/j.ijbiomac.2024.129618] [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: 07/14/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
Poor bioavailability, solubility, and absorption of berberine (Ber) limit its widespread application. Here, we formulated novel chitosan/pectin nanoparticles (NPs) loaded with Ber to address delivery problems and promote the anticancer properties of Ber in AGS gastric cancer cells. The ionic gelification method was used to synthesize NPs-Ber. Physicochemical characterization of NPs-Ber was performed using FE-SEM, DLS, PDI, ζ potential, and FTIR. The cytotoxic effects of NPs-Ber on AGS cells were evaluated using the MTT assay. Apoptosis and cell cycle arrest were examined by flow cytometry. The gene expression levels of miR-185-5p, KLF7, caspase-3, and DNMTs were determined using RT-qPCR. In addition, the 5-methylcytosine level in the genomic DNA was quantified using ELISA. FE-SEM images revealed a denser and more packed matrix for NPs-Ber, and FTIR analysis confirmed the formation of NPs-Ber. The size (550.39 nm), PDI (0.134), and ζ potential (-16.52 mV) confirmed the stability of the prepared NPs-Ber. NPs-Ber showed a continuous release pattern following the Korsmeyer-Peppas model such that 81.36 % of Ber was released from the formulation after 240 min. Compared to NPs and free Ber, NPs-Ber was found to possess higher anticancer activity in AGS cells. This result was indicated by the viability test and further clarified by augmented apoptosis and cell cycle arrest at the G0/G1 phase. The IC50 value of NP-Ber against AGS cells was significantly lower than those of free Ber and NPs. Interestingly, our results showed that NPs-Ber considerably changed the expression levels of miR-185-5p, KLF7, caspase-3, and DNMTs (DNMT1, 3A, and 3B) compared with unloaded NPs and free Ber. Additionally, 5-methylated cytosine (5-mC) levels in cells treated with NPs-Ber were significantly higher than those in cells treated with unloaded NPs or free Ber. In summary, the present study demonstrated that Ber encapsulation in NPs enhances its cytotoxic and epigenetic effects on AGS cells, suggesting the promising potential of NPs-Ber in GC therapy.
Collapse
Affiliation(s)
- Esmaeel Babaeenezhad
- Student Research Committee, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Rashidipour
- Student Research Committee, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran; Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Zohreh Jangravi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Moradi Sarabi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department of Biochemistry and Genetics, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Meliț LE, Mărginean CO, Borka Balas R. The Most Recent Insights into the Roots of Gastric Cancer. Life (Basel) 2024; 14:95. [PMID: 38255710 PMCID: PMC10817233 DOI: 10.3390/life14010095] [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: 11/29/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Helicobacter pylori (H. pylori) is the most common bacterial infection worldwide, usually being acquired during childhood, and its persistence into adulthood represents one of the main contributors of gastric carcinogenesis. Based on these statements, it would be of great importance to know if the most early premalignant transformation occurs in children or later since, this would enable the development of effective anti-tumorigenesis strategies. The interplay between H. pylori virulence factors, the host's responses modified by this infection, and the gastric microecology are complex and eventually lead to the development of gastric cancer in susceptible individuals. Several biomarkers were identified as major contributors of this long-lasting process, such as pepsinogens, gastrin 17, lipid-, glucose- and iron-metabolism parameters, immunity players, aberrant bacterial DNA methylation, H. pylori virulence factors, and hallmarks of gastric dysbiosis. Several of these biomarkers were also identified in children with H. pylori infection, independently of the presence of premalignant lesions, which were also proven to be present in a subgroup of H. pylori-infected children, especially those carrying extremely virulent strains. Therefore, the most incipient premalignant gastric changes might indeed occur early during childhood, opening a promising research gate for further studies to delineate the border between infection and cancer.
Collapse
Affiliation(s)
| | - Cristina Oana Mărginean
- Department of Pediatrics I, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, Târgu Mureș, Gheorghe Marinescu Street, No. 38, 540136 Târgu Mureș, Romania; (L.E.M.)
| | | |
Collapse
|
5
|
Tang SY, Zhou PJ, Meng Y, Zeng FR, Deng GT. Gastric cancer: An epigenetic view. World J Gastrointest Oncol 2022; 14:90-109. [PMID: 35116105 PMCID: PMC8790429 DOI: 10.4251/wjgo.v14.i1.90] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/17/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) poses a serious threat worldwide with unfavorable prognosis mainly due to late diagnosis and limited therapies. Therefore, precise molecular classification and search for potential targets are required for diagnosis and treatment, as GC is complicated and heterogeneous in nature. Accumulating evidence indicates that epigenetics plays a vital role in gastric carcinogenesis and progression, including histone modifications, DNA methylation and non-coding RNAs. Epigenetic biomarkers and drugs are currently under intensive evaluations to ensure efficient clinical utility in GC. In this review, key epigenetic alterations and related functions and mechanisms are summarized in GC. We focus on integration of existing epigenetic findings in GC for the bench-to-bedside translation of some pivotal epigenetic alterations into clinical practice and also describe the vacant field waiting for investigation.
Collapse
Affiliation(s)
- Si-Yuan Tang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Pei-Jun Zhou
- Cancer Research Institute, School of Basic Medicine Science, Central South University, School of Basic Medicine Science, Central South University 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Fu-Rong Zeng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guang-Tong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
6
|
Gómez A, Pato ML, Bujanda L, Sala N, Companioni O, Cosme Á, Tufano M, Hanly DJ, García N, Sanz-Anquela JM, Gisbert JP, López C, Elizalde JI, Cuatrecasas M, Andreu V, Paules MJ, Martín-Arranz MD, Ortega L, Poves E, Barrio J, Torres MÁ, Muñoz G, Ferrández Á, Ramírez-Lázaro MJ, Lario S, González CA, Esteller M, Berdasco M. Follow-Up Study Confirms the Presence of Gastric Cancer DNA Methylation Hallmarks in High-Risk Precursor Lesions. Cancers (Basel) 2021; 13:2760. [PMID: 34199386 PMCID: PMC8199626 DOI: 10.3390/cancers13112760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/31/2022] Open
Abstract
To adopt prevention strategies in gastric cancer, it is imperative to develop robust biomarkers with acceptable costs and feasibility in clinical practice to stratified populations according to risk scores. With this aim, we applied an unbiased genome-wide CpG methylation approach to a discovery cohort composed of gastric cancer (n = 24), and non-malignant precursor lesions (n = 64). Then, candidate-methylation approaches were performed in a validation cohort of precursor lesions obtained from an observational longitudinal study (n = 264), with a 12-year follow-up to identify repression or progression cases. H. pylori stratification and histology were considered to determine their influence on the methylation dynamics. As a result, we ascertained that intestinal metaplasia partially recapitulates patterns of aberrant methylation of intestinal type of gastric cancer, independently of the H. pylori status. Two epigenetically regulated genes in cancer, RPRM and ZNF793, consistently showed increased methylation in intestinal metaplasia with respect to earlier precursor lesions. In summary, our result supports the need to investigate the practical utilities of the quantification of DNA methylation in candidate genes as a marker for disease progression. In addition, the H. pylori-dependent methylation in intestinal metaplasia suggests that pharmacological treatments aimed at H. pylori eradication in the late stages of precursor lesions do not prevent epigenome reprogramming toward a cancer signature.
Collapse
Affiliation(s)
- Antonio Gómez
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain; (A.G.); (M.L.P.); (M.T.); (D.J.H.)
| | - Miguel L. Pato
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain; (A.G.); (M.L.P.); (M.T.); (D.J.H.)
- Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD, 20014 San Sebastián, Spain; (L.B.); (Á.C.)
| | - Núria Sala
- Unit of Nutrition, Environment and Cancer, Institut Català d’Oncología, 08908 Barcelona, Spain; (N.S.); (O.C.); (N.G.); (C.A.G.)
- Translational Research Laboratory, Catalan Institute of Oncology (ICO)-IDIBELL, 08908 Barcelona, Spain
| | - Osmel Companioni
- Unit of Nutrition, Environment and Cancer, Institut Català d’Oncología, 08908 Barcelona, Spain; (N.S.); (O.C.); (N.G.); (C.A.G.)
| | - Ángel Cosme
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD, 20014 San Sebastián, Spain; (L.B.); (Á.C.)
| | - Martina Tufano
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain; (A.G.); (M.L.P.); (M.T.); (D.J.H.)
| | - David J. Hanly
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain; (A.G.); (M.L.P.); (M.T.); (D.J.H.)
- Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| | - Nadia García
- Unit of Nutrition, Environment and Cancer, Institut Català d’Oncología, 08908 Barcelona, Spain; (N.S.); (O.C.); (N.G.); (C.A.G.)
- Translational Research Laboratory, Catalan Institute of Oncology (ICO)-IDIBELL, 08908 Barcelona, Spain
| | - José Miguel Sanz-Anquela
- Department of Pathology, Hospital Universitario Príncipe de Asturias, 28805 Alcalá de Henares, Spain;
| | - Javier P. Gisbert
- Department of Gastroenterology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (J.P.G.); (C.L.)
| | - Consuelo López
- Department of Gastroenterology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (J.P.G.); (C.L.)
| | - José Ignacio Elizalde
- Department of Gastroenterology, Hospital Clínic de Barcelona, IDIBAPS and CIBEREHD, 08036 Barcelona, Spain;
| | - Miriam Cuatrecasas
- Department of Pathology, Hospital Clínic de Barcelona, IDIBAPS and CIBEREHD, 08036 Barcelona, Spain;
| | - Victoria Andreu
- Department of Gastroenterology, Hospital de Viladecans, 08840 Barcelona, Spain;
| | - María José Paules
- Department of Pathology, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain;
| | - María Dolores Martín-Arranz
- Department of Gastroenterology, Hospital Universitario La Paz, Instituto de Investigación Sanitaria La Paz (IdiPaz), 28046 Madrid, Spain;
| | - Luis Ortega
- Department of Gastroenterology, Hospital Clínico San Carlos, 28040 Madrid, Spain;
| | - Elvira Poves
- Department of Gastroenterology, Hospital Universitario Príncipe de Asturias, 28805 Alcalá de Henares, Spain;
| | - Jesús Barrio
- Department of Gastroenterology, Hospital Universitario Río Hortega, 47012 Valladolid, Spain;
| | - María Ángeles Torres
- Department of Pathology, Hospital Universitario Río Hortega, 47012 Valladolid, Spain;
| | - Guillermo Muñoz
- Department of Gastroenterology, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain; (G.M.); (Á.F.)
| | - Ángel Ferrández
- Department of Gastroenterology, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain; (G.M.); (Á.F.)
| | - María José Ramírez-Lázaro
- Department of Medicine, Digestive Diseases Service, Institut Universitari Parc Taulí, 08201 Sabadell, Spain; (M.J.R.-L.); (S.L.)
| | - Sergio Lario
- Department of Medicine, Digestive Diseases Service, Institut Universitari Parc Taulí, 08201 Sabadell, Spain; (M.J.R.-L.); (S.L.)
| | - Carlos A González
- Unit of Nutrition, Environment and Cancer, Institut Català d’Oncología, 08908 Barcelona, Spain; (N.S.); (O.C.); (N.G.); (C.A.G.)
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer and Leukemia Epigenetics and Biology Program (PEBCL), Josep Carreras Leukaemia Research Institute (IJC), 08916 Barcelona, Spain;
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - María Berdasco
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain; (A.G.); (M.L.P.); (M.T.); (D.J.H.)
- Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukaemia Research Institute, 08916 Barcelona, Spain
| |
Collapse
|
7
|
Bakhti SZ, Latifi-Navid S, Gholizade Tobnagh S, Yazdanbod K, Yazdanbod A. Which genotype of Helicobacter pylori—cagA or cagE—Is better associated with gastric Cancer risk? Lessons from an extremely high-risk area in Iran. INFECTION, GENETICS AND EVOLUTION 2020; 85:104431. [DOI: 10.1016/j.meegid.2020.104431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
|
8
|
Youssefi M, Ghazvini K, Farsiani H, Tafaghodi M, Keikha M. A systematic review and meta-analysis of outcomes of infection with Helicobacter pylori dupA+ strains in Iranian patients. GENE REPORTS 2020; 19:100650. [DOI: 10.1016/j.genrep.2020.100650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
9
|
Ali H, Sharif M, Yasmin M, Rehmani MH, Riaz F. A survey of feature extraction and fusion of deep learning for detection of abnormalities in video endoscopy of gastrointestinal-tract. Artif Intell Rev 2019. [DOI: 10.1007/s10462-019-09743-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
10
|
Hoffman RM. Is the Hoffman Effect for Methionine Overuse Analogous to the Warburg Effect for Glucose Overuse in Cancer? Methods Mol Biol 2019; 1866:273-278. [PMID: 30725423 DOI: 10.1007/978-1-4939-8796-2_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
The general cancer-specific metabolic defect of methionine (MET) dependence is due to MET overuse for aberrant transmethylation reactions. The excess use of MET for aberrant transmethylation reactions apparently diverts methyl groups from DNA. The resulting global DNA hypomethylation is also a general phenomenon in cancer and leads to unstable genomes and aneuploid karyotypes. The excessive and aberrant use of MET in cancer is readily observed in [11C]-MET-PET imaging, where high uptake of [11C]-MET results in a very strong and selective tumor signal compared to normal tissue background for brain cancer and possibly other cancers. [11C]-MET is superior to [18C]-fluorodeoxyglucose (FDG) for PET imaging, suggesting that MET overuse in cancer ("Hoffman effect") is greater than glucose overuse in cancer ("Warburg effect").
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA.
- Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
11
|
Ding N, Maiuri AR, O'Hagan HM. The emerging role of epigenetic modifiers in repair of DNA damage associated with chronic inflammatory diseases. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2019; 780:69-81. [PMID: 31395351 PMCID: PMC6690501 DOI: 10.1016/j.mrrev.2017.09.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022]
Abstract
At sites of chronic inflammation epithelial cells are exposed to high levels of reactive oxygen species (ROS), which can contribute to the initiation and development of many different human cancers. Aberrant epigenetic alterations that cause transcriptional silencing of tumor suppressor genes are also implicated in many diseases associated with inflammation, including cancer. However, it is not clear how altered epigenetic gene silencing is initiated during chronic inflammation. The high level of ROS at sites of inflammation is known to induce oxidative DNA damage in surrounding epithelial cells. Furthermore, DNA damage is known to trigger several responses, including recruitment of DNA repair proteins, transcriptional repression, chromatin modifications and other cell signaling events. Recruitment of epigenetic modifiers to chromatin in response to DNA damage results in transient covalent modifications to chromatin such as histone ubiquitination, acetylation and methylation and DNA methylation. DNA damage also alters non-coding RNA expression. All of these alterations have the potential to alter gene expression at sites of damage. Typically, these modifications and gene transcription are restored back to normal once the repair of the DNA damage is completed. However, chronic inflammation may induce sustained DNA damage and DNA damage responses that result in these transient covalent chromatin modifications becoming mitotically stable epigenetic alterations. Understanding how epigenetic alterations are initiated during chronic inflammation will allow us to develop pharmaceutical strategies to prevent or treat chronic inflammation-induced cancer. This review will focus on types of DNA damage and epigenetic alterations associated with chronic inflammatory diseases, the types of DNA damage and transient covalent chromatin modifications induced by inflammation and oxidative DNA damage and how these modifications may result in epigenetic alterations.
Collapse
Affiliation(s)
- Ning Ding
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA
| | - Ashley R Maiuri
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA
| | - Heather M O'Hagan
- Medical Sciences Program, School of Medicine, Indiana University, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
12
|
Recombinant Methioninase as a DNA Demethylation Agent. Methods Mol Biol 2019. [PMID: 30725424 DOI: 10.1007/978-1-4939-8796-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
This chapter reviews the effect of methionine (MET) restriction, via treatment with recombinant methioninase (rMETase), on DNA methylation of cancer cells. CCRF-CEM human cancer cells were treated with rMETase under subcytotoxic conditions. The rMETase-treated cells contained significantly lower levels of genomic methylated DNA than did untreated control cells. DNA methylation was measured by incorporation of the methyl group of [3H]methyl-S-adenosylmethionine into DNA and by methylation-sensitive arbitrarily-primed PCR. DNA hypomethylation effected by rMETase was of similar extent to that effected by treatment of the cells with the DNA methyltransferase inhibitor 5-azacytidine.
Collapse
|
13
|
Lu JJ, Yuan Z. Application of DNA methylation in early diagnosis and treatment of pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2019; 27:13-19. [DOI: 10.11569/wcjd.v27.i1.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant gastrointestinal tumors, characterized by a poor prognosis. Most of the patients have an advanced disease at the time of diagnosis and lose the opportunity of radical surgery, resulting in a 5-year survival rate of less than 5%. Circulating tumor DNA, whose concentration in plasma of patients with pancreatic adenocarcinoma is higher than that in health controls, carries specific gene mutation and aberrant DNA methylation. Epigenetic change is one of the important characteristics of cell carcinogenesis. DNA methylation is an early event in tumorigenesis, which is more helpful for early diagnosis than gene mutation and can be observed in each stage of PC. Therefore, the detection of aberrant DNA methylation in the promoter region in patients with PC may be a non-invasive method for early cancer detection, predicting prognosis, and monitoring recurrence. In the present review, we discuss the recent advances in the study of DNA methylation in the early diagnosis of PC, and the potential application value in the treatment of PC.
Collapse
Affiliation(s)
- Jia-Jun Lu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhou Yuan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
14
|
Fluctuations of epigenetic regulations in human gastric Adenocarcinoma: How does it affect? Biomed Pharmacother 2019; 109:144-156. [DOI: 10.1016/j.biopha.2018.10.094] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022] Open
|
15
|
Hoffman RM, Stern PH, Coalson DW, Douglas Wallace C, Erbe RW. Altered Methionine Metabolism in Cancer Cells. Methods Mol Biol 2019; 1866:13-26. [PMID: 30725404 DOI: 10.1007/978-1-4939-8796-2_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Many different types of cancer cells have been shown to be methionine (MET) dependent. Cancer cells, unlike normal cells, grow poorly or not at all when MET is restricted. Cancer cells have an elevated requirement for exogenous MET for growth, despite high levels of endogenous synthesis. This requirement reflects increased utilization of MET by cancer cells, analogous to increased utilization glucose by cancer cells (Warburg effect). To answer the critical question of whether MET-dependent cancer cells synthesize normal amounts of MET, we determined the levels of MET, S-adenosylmethionine (AdoMET), and S-adenosylhomocysteine (AdoHCY) that were synthesized by MET-dependent cancer cells under conditions of MET restriction. We demonstrated that MET-dependent cells synthesize a normal amount of endogenously synthesized MET but are still deficient in AdoMET. In contrast, exogenously supplied MET results in normal AdoMET levels. The ratio of AdoMET to AdoHCY is low in MET-dependent cells growing in MET-restricted medium but is normal when MET is supplied. Under conditions of MET restriction, the low AdoMET/AdoHCY ratio probably limits proliferation of MET-dependent cancer cells. The amount of free MET is also low in MET-dependent cancer cells under MET restriction. The elevated MET requirement for cancer cells may be due to enhanced overall rates of transmethylation compared to normal human cells. Thus, MET-dependent cancer cells have low levels of free MET, low levels of AdoMET, and elevated levels of AdoHCY under conditions of MET restriction probably due to overuse of MET for transmethylation reactions ("Hoffman effect"), thereby blocking cellular proliferation.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA. .,Department of Surgery, University of California, San Diego, CA, USA.
| | | | - Dennis W Coalson
- Anesthesia and Critical Care, University of Chicago Medicine, Chicago, IL, USA
| | | | - Richard W Erbe
- Pediatrics and Medicine, University at Buffalo, Buffalo, NY, USA.,Division of Genetics, The Women and Children's Hospital of Buffalo, Buffalo, NY, USA
| |
Collapse
|
16
|
Zhong F, Zhu M, Gao K, Xu P, Yang H, Hu D, Cui D, Wang M, Xie X, Wei Y, Zhang H, Du H. Low expression of the long non-coding RNA NR_026827 in gastric cancer. Am J Transl Res 2018; 10:2706-2711. [PMID: 30210707 PMCID: PMC6129519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 07/16/2018] [Indexed: 06/08/2023]
Abstract
AIM The contribution of long non-coding RNAs (lncRNAs) to gastric cancer associated with Helicobacter pylori (H. pylori) infection remains largely unknown. Therefore, the present study aimed to investigate the expression of a lncRNA NR_026827 in gastric epithelial cells infected with H. pylori, and demonstrate its expression characteristic in gastric cancer. MATERIALS AND METHODS Gastric epithelial cell line cells, GES-1, were cultured and infected with H. pylori. A microarray was used to analyze the lncRNA profile of gastric epithelial cells. Eighty fresh gastric cancer tissues and the paired adjacent non-cancerous tissue samples were randomly selected from patients. The expression of the lncRNA NR_026827 was investigated using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS The expression of several lncRNAs was significantly altered in GES-1 cells following infection with H. pylori. Of these lncRNAs, NR_026827 was dramatically down-regulated in GES-1 cells infected with H. pylori. In addition, the expression of NR_026827 was decreased in gastric cancer tissues in comparison to the corresponding adjacent non-cancerous tissues. Moreover, the expression of NR_026827 did not change significantly in different gastric cancer stages. CONCLUSION The lncRNA, NR_026827, is down-regulated in all stages of gastric cancer associated with H. pylori infection and could represent a potential biomarker for the diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Fengyun Zhong
- Department of General Surgery, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Meili Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Kaijian Gao
- Department of General Surgery, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Ping Xu
- Department of Clinical Laboratory, The Fifth People’s Hospital of SuzhouSuzhou 215007, Jiangsu, PR China
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Dawei Cui
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang UniversityHangzhou 310006, Zhejiang, PR China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Xiaofang Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Yan Wei
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, PR China
| |
Collapse
|
17
|
Katoh M. Multi‑layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β‑catenin signaling activation (Review). Int J Mol Med 2018; 42:713-725. [PMID: 29786110 PMCID: PMC6034925 DOI: 10.3892/ijmm.2018.3689] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
β-catenin/CTNNB1 is an intracellular scaffold protein that interacts with adhesion molecules (E-cadherin/CDH1, N-cadherin/CDH2, VE-cadherin/CDH5 and α-catenins), transmembrane-type mucins (MUC1/CD227 and MUC16/CA125), signaling regulators (APC, AXIN1, AXIN2 and NHERF1/EBP50) and epigenetic or transcriptional regulators (BCL9, BCL9L, CREBBP/CBP, EP300/p300, FOXM1, MED12, SMARCA4/BRG1 and TCF/LEF). Gain-of-function CTTNB1 mutations are detected in bladder cancer, colorectal cancer, gastric cancer, liver cancer, lung cancer, pancreatic cancer, prostate cancer and uterine cancer, whereas loss-of-function CTNNB1 mutations are also detected in human cancer. ABCB1, ALDH1A1, ASCL2, ATF3, AXIN2, BAMBI, CCND1, CD44, CLDN1, CTLA4, DKK1, EDN1, EOMES, FGF18, FGF20, FZD7, IL10, JAG1, LEF1, LGR5, MITF, MSX1, MYC, NEUROD1, NKD1, NODAL, NOTCH2, NOTUM, NRCAM, OPN, PAX3, PPARD, PTGS2, RNF43, SNAI1, SP5, TCF7, TERT, TNFRSF19, VEGFA and ZNRF3 are representative β-catenin target genes. β-catenin signaling is involved in myofibroblast activation and subsequent pulmonary fibrosis, in addition to other types of fibrosis. β-catenin and NF-κB signaling activation are involved in field cancerization in the stomach associated with Helicobacter pylori (H. pylori) infection and in the liver associated with hepatitis C virus (HCV) infection and other etiologies. β-catenin-targeted therapeutics are functionally classified into β-catenin inhibitors targeting upstream regulators (AZ1366, ETC-159, G007-LK, GNF6231, ipafricept, NVP-TNKS656, rosmantuzumab, vantictumab, WNT-C59, WNT974 and XAV939), β-catenin inhibitors targeting protein-protein interactions (CGP049090, CWP232228, E7386, ICG-001, LF3 and PRI-724), β-catenin inhibitors targeting epigenetic regulators (PKF118-310), β-catenin inhibitors targeting mediator complexes (CCT251545 and cortistatin A) and β-catenin inhibitors targeting transmembrane-type transcriptional outputs, including CD44v6, FZD7 and LGR5. Eradicating H. pylori and HCV is the optimal approach for the first-line prevention of gastric cancer and hepatocellular carcinoma (HCC), respectively. However, β-catenin inhibitors may be applicable for the prevention of organ fibrosis, second-line HCC prevention and treating β-catenin-driven cancer. The multi-layered prevention and treatment strategy of β-catenin-related human diseases is necessary for the practice of personalized medicine and implementation of precision medicine.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Chuo Ward, Tokyo 104‑0045, Japan
| |
Collapse
|
18
|
Pormohammad A, Ghotaslou R, Leylabadlo HE, Nasiri MJ, Dabiri H, Hashemi A. Risk of gastric cancer in association with Helicobacter pylori different virulence factors: A systematic review and meta-analysis. Microb Pathog 2018; 118:214-219. [PMID: 29510208 DOI: 10.1016/j.micpath.2018.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION It has been proposed that specific analysis of Helicobacter pylori virulence factors can be suitable for predicting of post H. pylori infection disorders like gastric cancer (GC). The present study was designed to evaluate the association between different virulence factors of H. pylori and GC. METHODS Studies investigated the association between virulence factors of H. pylori and GC were collected from the several databases. All analysis was performed by Comprehensive Meta-Analysis V2.2 software (Biostat, Englewood, NJ, USA). RESULTS Based on a comprehensive literature search, 25 eligible studies were included for meta-analyses. Infection with cagA- and vacA s1m1-positive H. pylori strains were significantly associated with increased risk of GC (OR of [2.82 (95% CI 1.96-4.06), P < 0.001]) and ([1.75 (95% CI 1.04-2.96), P 0.034)], respectively. CONCLUSIONS Infection by H. pylori strains with positive vacA s1m1 and the cagA genes can significantly increase the risk of GC. The association between the vacA s1m1 and the cagA and GC, suggests that screening of these genes may be helpful for identifying populations at higher risk for GC.
Collapse
Affiliation(s)
- Ali Pormohammad
- Student Research Committee, Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Ghotaslou
- Infectious and Tropical Diseases Research Center and Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Dabiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Fukuyama T, Futawatari N, Yamamura R, Yamazaki T, Ichiki Y, Ema A, Ushiku H, Nishi Y, Takahashi Y, Otsuka T, Yamazaki H, Koizumi W, Yasumoto K, Kobayashi N. Expression of KK-LC-1, a cancer/testis antigen, at non-tumour sites of the stomach carrying a tumour. Sci Rep 2018; 8:6131. [PMID: 29666402 PMCID: PMC5904109 DOI: 10.1038/s41598-018-24514-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/27/2018] [Indexed: 02/07/2023] Open
Abstract
Kita-Kyushu lung cancer antigen-1 (KK-LC-1) is a cancer/testis antigen (CTA) and predominant target for cancer immunotherapy. Our previous study indicated that KK-LC-1 was expressed in 82% of gastric cancers, and also in 79% of early stage of gastric cancers, with a correlation to Helicobacter pylori (H. pylori) infection. In addition, we found that KK-LC-1 was occasionally expressed at non-tumour sites of stomachs carrying tumours. Here, we investigated the characteristics of KK-LC-1 expression at non-tumour sites and the clinical utility of these phenomena. The gene expression of KK-LC-1 was detected at the non-tumour sites including pyloric glands. The most detectable corpus/gland subset had a KK-LC-1 expression rate of 77% in the pyloric gland of the lower corpus where H. pylori preferentially exists. KK-LC-1 expression rates were 67% or 32% with or without intestinal metaplasia, which also induced by H. pylori, respectively. Consequently, KK-LC-1 would be detected at the pre-cancerous condition of the stomach, and may be a useful marker to predict gastric cancer.
Collapse
Affiliation(s)
- Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan.
| | - Nobue Futawatari
- Department of Surgery, Sagamihara National Hospital, Sagamihara, Japan
| | - Rui Yamamura
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Taiga Yamazaki
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Yoshinobu Ichiki
- Second Department of Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akira Ema
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Hideki Ushiku
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Japan.,Division of Surgery, Kitasato University Medical Center, Kitamoto, Japan
| | - Yatsushi Nishi
- Division of Surgery, Kitasato University Medical Center, Kitamoto, Japan
| | - Yoshihito Takahashi
- Department of Surgery, School of Medicine, Kitasato University, Sagamihara, Japan.,Division of Surgery, Kitasato University Medical Center, Kitamoto, Japan
| | - Toshikazu Otsuka
- Division of Gastroenterology, Kitasato University Medical Center, Kitamoto, Japan
| | - Hitoshi Yamazaki
- Division of Pathology, Kitasato University Medical Center, Kitamoto, Japan
| | - Wasaburo Koizumi
- Department of Gastroenterology, School of Medicine, Kitasato University, Sagamihara, Japan
| | | | - Noritada Kobayashi
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| |
Collapse
|
20
|
Pirini F, Noazin S, Jahuira-Arias MH, Rodriguez-Torres S, Friess L, Michailidi C, Cok J, Combe J, Vargas G, Prado W, Soudry E, Pérez J, Yudin T, Mancinelli A, Unger H, Ili-Gangas C, Brebi-Mieville P, Berg DE, Hayashi M, Sidransky D, Gilman RH, Guerrero-Preston R. Early detection of gastric cancer using global, genome-wide and IRF4, ELMO1, CLIP4 and MSC DNA methylation in endoscopic biopsies. Oncotarget 2018; 8:38501-38516. [PMID: 28418867 PMCID: PMC5503549 DOI: 10.18632/oncotarget.16258] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/24/2017] [Indexed: 12/15/2022] Open
Abstract
Clinically useful molecular tools to triage gastric cancer patients are not currently available. We aimed to develop a molecular tool to predict gastric cancer risk in endoscopy-driven biopsies obtained from high-risk gastric cancer clinics in low resource settings. We discovered and validated a DNA methylation biomarker panel in endoscopic samples obtained from 362 patients seen between 2004 and 2009 in three high-risk gastric cancer clinics in Lima, Perú, and validated it in 306 samples from the Cancer Genome Atlas project (“TCGA”). Global, epigenome wide and gene-specific DNA methylation analyses were used in a Phase I Biomarker Development Trial to identify a continuous biomarker panel that combines a Global DNA Methylation Index (GDMI) and promoter DNA methylation levels of IRF4, ELMO1, CLIP4 and MSC. We observed an inverse association between the GDMI and histological progression to gastric cancer, when comparing gastritis patients without metaplasia (mean = 5.74, 95% CI, 4.97−6.50), gastritis patients with metaplasia (mean = 4.81, 95% CI, 3.77−5.84), and gastric cancer cases (mean = 3.38, 95% CI, 2.82−3.94), respectively (p < 0.0001). Promoter methylation of IRF4 (p < 0.0001), ELMO1 (p < 0.0001), CLIP4 (p < 0.0001), and MSC (p < 0.0001), is also associated with increasing severity from gastritis with no metaplasia to gastritis with metaplasia and gastric cancer. Our findings suggest that IRF4, ELMO1, CLIP4 and MSC promoter methylation coupled with a GDMI>4 are useful molecular tools for gastric cancer risk stratification in endoscopic biopsies.
Collapse
Affiliation(s)
- Francesca Pirini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Sassan Noazin
- The Johns Hopkins University, Bloomberg School of Public Health, Department of International Health, Baltimore, MD, USA
| | - Martha H Jahuira-Arias
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Sebastian Rodriguez-Torres
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Leah Friess
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Christina Michailidi
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Jaime Cok
- Hospital Nacional Cayetano Heredia, Pathology Department, Lima, Perú
| | - Juan Combe
- Instituto Nacional de Enfermedades Neoplásicas, Gastroenterology Department, Lima, Perú
| | - Gloria Vargas
- Hospital Nacional Arzobispo Loayza, Gastroenterology Department, Lima, Perú
| | - William Prado
- Hospital Nacional Dos de Mayo, Gastroenterology Department, Lima, Perú
| | - Ethan Soudry
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Jimena Pérez
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Tikki Yudin
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Andrea Mancinelli
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Helen Unger
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Carmen Ili-Gangas
- Laboratory of Molecular Pathology, Department of Pathological Anatomy, School of Medicine, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Priscilla Brebi-Mieville
- Laboratory of Molecular Pathology, Department of Pathological Anatomy, School of Medicine, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Translational Medicine - Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Douglas E Berg
- Washington University Medical School, Department of Molecular Microbiology, St Louis, MO, USA.,University of California San Diego, Department of Medicine, La Jolla, CA, USA
| | - Masamichi Hayashi
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - David Sidransky
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA
| | - Robert H Gilman
- The Johns Hopkins University, Bloomberg School of Public Health, Department of International Health, Baltimore, MD, USA.,Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Rafael Guerrero-Preston
- The Johns Hopkins University, School of Medicine, Otolaryngology Department, Head and Neck Cancer Research Division, Baltimore, MD, USA.,University of Puerto Rico School of Medicine, Department of Obstetrics and Gynecology, San Juan, Puerto Rico
| |
Collapse
|
21
|
Abstract
Gastric cancer is a deadly malignancy afflicting close to a million people worldwide. Patient survival is poor and largely due to late diagnosis and suboptimal therapies. Disease heterogeneity is a substantial obstacle, underscoring the need for precision treatment strategies. Studies have identified different subgroups of gastric cancer displaying not just genetic, but also distinct epigenetic hallmarks. Accumulating evidence suggests that epigenetic abnormalities in gastric cancer are not mere bystander events, but rather promote carcinogenesis through active mechanisms. Epigenetic aberrations, induced by pathogens such as Helicobacter pylori, are an early component of gastric carcinogenesis, probably preceding genetic abnormalities. This Review summarizes our current understanding of the gastric cancer epigenome, highlighting key advances in recent years in both tumours and pre-malignant lesions, made possible through targeted and genome-wide technologies. We focus on studies related to DNA methylation and histone modifications, linking these findings to potential therapeutic opportunities. Lessons learned from the gastric cancer epigenome might also prove relevant for other gastrointestinal cancers.
Collapse
|
22
|
Zeng XQ, Wang J, Chen SY. Methylation modification in gastric cancer and approaches to targeted epigenetic therapy (Review). Int J Oncol 2017; 50:1921-1933. [DOI: 10.3892/ijo.2017.3981] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/22/2017] [Indexed: 11/06/2022] Open
|
23
|
Hoffman RM. Is DNA methylation the new guardian of the genome? Mol Cytogenet 2017; 10:11. [PMID: 28396696 PMCID: PMC5381125 DOI: 10.1186/s13039-017-0314-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/28/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND It has been known for more than 100 years that aneuploidy is an essence of cancer. The question is what keeps the genome stable, thereby preventing aneuploidy. For the past 25 years, it has been proposed that p53 is the "guardian of the genome." However, it has been shown that inactivation of p53 does not cause aneuploidy. Another essence of cancer is global DNA hypomethylation, which causes destabilization of the genome and subsequent aneupoloidy. Yet, another essence of cancer is excessive use of methionine, resulting in methionine dependence. Methionine dependence is due to possible "metabolic reprogramming" due to carcinogens, including chemical agents and infectious organisms, such as Helicobacter pylori, that result in altered and excessive transmethylation in cancer cells. Cancer cells appear to have a "methyl-sink" whereby methyl groups are diverted from DNA. CONCLUSION DNA hypomethylation destabilizes the genome, leading to aneuploidy and subsequent selection and speciation into autonomous cancers, leading to the conclusion that DNA methylation is the "guardian of the genome."
Collapse
Affiliation(s)
- Robert M. Hoffman
- Anti Cancer Inc, 7917 Ostrow Street, San Diego, 92111 CA USA
- Department of Surgery, University of California, San Diego, CA USA
| |
Collapse
|
24
|
Abstract
We propose here a hypothesis of the cause of cancer that brings together fundamental changes in methyl-group metabolism resulting in methionine dependence and global DNA hypomethylation which destabilizes the genome leading to aneuploid karyotypes which evolve and stabilize into autonomous cancer. Experimental support for this hypothesis is that methioine dependence is a general metabolic defect in caner. Methionine dependence is due to excess use of methionene for aberrant transmethylation reactions that apparently divert methyl groups from DNA. The resulting global DNA hypomethylation is also a general phenomena in cancer. Global hypomethylation leads to an unstable genomes and aneuploid karyotypes, another general phenomena in cancer. The excessive and aberrant use of methionine in cancer is strongly observed in [11C]methionine PET imaging, where high uptake of [11C]methionine results in a very strong and selective tumor signal compared with normal tissue background. [11C]methionine is superior to [18C] fluorodeoxyglucose (FDG)-PET for PET imaging, suggesting methionine dependence is more tumor-specific than glucose dependence.
Collapse
Affiliation(s)
- Robert M Hoffman
- a AntiCancer Inc. , San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| |
Collapse
|
25
|
Zhuo C, Li Q, Wu Y, Li Y, Nie J, Li D, Peng J, Lian P, Li B, Cai G, Li X, Cai S. LINE-1 hypomethylation in normal colon mucosa is associated with poor survival in Chinese patients with sporadic colon cancer. Oncotarget 2016; 6:23820-36. [PMID: 26172297 PMCID: PMC4695154 DOI: 10.18632/oncotarget.4450] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/19/2015] [Indexed: 12/20/2022] Open
Abstract
Genetic and epigenetic pathways are not independent in colorectal cancer (CRC) carcinogenesis. We aimed to determine the influence of various molecular features on Chinese patients' colon cancer-specific survival (CCSS). Various genetic and epigenetic modifications were detected in paired tumor and normal mucosa tissue samples. The prognostic variables regarding patient CCSS were determined. Overall, 127 patients, including 83 males and 44 females, completed a median follow-up of 65 (3–85) months. A mean LINE-1 methylation rate of 64.62% (range, 9.45–86.93) was observed. Hypermethylation at the hMLH1 gene promoter was detected in 26 (20.47%) patients. KRAS was mutated in 52 (40.94%) patients. Sixteen (12.60%) patients were confirmed as microsatellite instability (MSI)-High, and 76 (59.84%) were found to have loss of heterozygosity at 18q. The LINE-1 methylation level, MSI status, perineural invasion and distant metastases were confirmed as independent prognostic factors for patient CCSS. A stratified survival analysis further revealed that certain subgroups of patients with LINE-1 hypomethylation had significantly worse survival (all p < 0.05). Our data revealed that both genetic and epigenetic abnormalities can concurrently exist during colonic tumorigenesis. As a global epigenetic change, LINE-1 hypomethylation in normal colon mucosa might be associated with a worse outcome in certain Chinese patients with colon cancer.
Collapse
Affiliation(s)
- Changhua Zhuo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Surgical Oncology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian Medical University, Fuzhou 350014, People's Republic of China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yuchen Wu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yiwei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Jia Nie
- Key Laboratory of Molecular Virology & Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Peng Lian
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Bin Li
- Key Laboratory of Molecular Virology & Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
26
|
Abstract
Oxidative stress has a significant impact on the development and progression of common human pathologies, including cancer, diabetes, hypertension and neurodegenerative diseases. Increasing evidence suggests that oxidative stress globally influences chromatin structure, DNA methylation, enzymatic and non-enzymatic post-translational modifications of histones and DNA-binding proteins. The effects of oxidative stress on these chromatin alterations mediate a number of cellular changes, including modulation of gene expression, cell death, cell survival and mutagenesis, which are disease-driving mechanisms in human pathologies. Targeting oxidative stress-dependent pathways is thus a promising strategy for the prevention and treatment of these diseases. We summarize recent research developments connecting oxidative stress and chromatin regulation.
Collapse
Affiliation(s)
- Sarah Kreuz
- King Abdullah University of Science & Technology (KAUST), Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia
| | - Wolfgang Fischle
- King Abdullah University of Science & Technology (KAUST), Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
27
|
Baba Y, Ishimoto T, Kurashige J, Iwatsuki M, Sakamoto Y, Yoshida N, Watanabe M, Baba H. Epigenetic field cancerization in gastrointestinal cancers. Cancer Lett 2016; 375:360-366. [PMID: 26971491 DOI: 10.1016/j.canlet.2016.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 02/06/2023]
Abstract
Epigenetic alterations, including aberrant DNA methylation, play an important role in human cancer development. Importantly, epigenetic alterations are reversible and can be targets for therapy or chemoprevention for various types of human cancers. A field for cancerization, or a field defect, is formed by the accumulation of genetic and/or epigenetic alterations in normal-appearing tissues and can correlate with risk of cancer development. Thus, a better understanding of epigenetic field cancerization may represent a useful translational opportunity for cancer risk assessment, including previous history and exposure to carcinogenic factors, and for cancer prevention. In this article, we summarize current knowledge regarding epigenetic field cancerization and its clinical implications in gastrointestinal cancers, including colorectal cancer, gastric cancer and esophageal cancer.
Collapse
Affiliation(s)
- Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Junji Kurashige
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Japan.
| |
Collapse
|