1
|
Wang Z, Li T, Sun M, Liu N, Zhang H, Feng Z, Lei N. Metabolomics- and Proteomics-Based Disease Diagnostic Classifier Model for the Prediction and Diagnosis of Colorectal Carcinoma. J Proteome Res 2025. [PMID: 40108892 DOI: 10.1021/acs.jproteome.5c00010] [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: 03/22/2025]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is a leading cause of cancer-related deaths globally. Diagnostic biomarkers are essential for risk stratification and early detection, potentially enhancing patient survival. Our study aimed to explore the potential biomarkers of CRC at the protein and metabolic levels. METHODS Blood serum from CRC patients and healthy controls was analyzed using metabolomic and proteomic techniques. A conjoint analysis was conducted, and samples were split into training and validation sets (7:3 ratio) to develop and evaluate a disease diagnosis classifier model. Immunohistochemistry (IHC) analyses were conducted to validate the results. RESULTS We identified 631 differential metabolites and 61 differentially expressed proteins (DEPs) in CRC, involved in pathways such as arginine and proline metabolism, central carbon metabolism in cancer, and signaling pathways including TGF-β, mTOR, PI3K-Akt, and others. Key proteins (CILP2, SLC3A2, EXTL2, hydroxypyruvate isomerase (HYI), ENPEP, LRG1, CTSS, thyrotropin-releasing hormone-degrading ectoenzyme (TRHDE), SELE, and HSPA1A) showed significant expression differences between CRC patients and controls. IHC results showed that compared with the paracancerous tissues, the expression of CILP2, EXTL2, and HYI was significantly downregulated in the CRC tissues (P < 0.05). The classifier model, comprising l-arginine, Harden-Young ester, l-aspartic acid, oxoglutaric acid, l-proline, octopine, l-valine, and progesterone, achieved AUC values of 0.998 and 0.914 in training and validation data sets, respectively. CONCLUSIONS The identified metabolites and DEPs are promising CRC biomarkers. The developed classifier model based on eight metabolites demonstrates high accuracy for CRC assessment and diagnosis.
Collapse
Affiliation(s)
- Zhaorui Wang
- Translational Medicine Research Center, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Tianyuan Li
- Department of Gastroenterology, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Mengyao Sun
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Na Liu
- Translational Medicine Research Center, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Haozhe Zhang
- Translational Medicine Research Center, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Zhikun Feng
- Department of Pathology, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, Henan 450000, China
| | - Ningjing Lei
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
2
|
Li JH, Tang Y, Wang J, Wei XF, Wang N, Wang JW, Lyu H, Jiang XM, Liu HH, Wang K. A Clinical Predictive Model Based on SOCS3 Promoter Methylation to Predict the Prognosis of Acute-on-Chronic Hepatitis B Liver Failure. J Inflamm Res 2025; 18:3741-3756. [PMID: 40109653 PMCID: PMC11920633 DOI: 10.2147/jir.s506050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Purpose The study aimed to quantitatively detect the suppressors of cytokine signaling (SOCS) 3 promoter methylation levels, investigate the relationship between SOCS3 methylation and gene expression, and construct a prognosis prediction model combined with clinical indicators for Acute-on-chronic Hepatitis B Liver Failure (ACHBLF). Methods A total of 135 ACHBLF patients were enrolled and randomly divided into the training cohort and validation cohort. The SOCS3 mRNA and promoter methylation in peripheral blood mononuclear cells (PBMCs) of ACHBLF patients were quantitative measured. A clinical prediction model was established based on SOCS3 promoter methylation and clinical indicators. The prediction model was evaluated by the area under the receiver operating characteristic curve, the Hosmer-Lemeshow (H-L) goodness-of-fit test, and decision curve analysis. Results In this study, compared with ACHBLF survivals, SOCS3 showed lower mRNA levels and higher methylation levels in ACHBLF non-survivals. The SOCS3 methylation rates were negatively correlated with SOCS3 mRNA levels. PT-INR, IL-6, and percentage of the methylation reference (PMR) value (SOCS3) were used to establish a clinical model for predicting ACHBLF patients' prognosis. The results of AUC, the Hosmer-Lemeshow (H-L) goodness-of-fit test and decision curve analysis (DCA) showed that the prediction model had good clinical applicability. The prediction model was visualized. Conclusion A prognosis prediction model for ACHBLF was developed based on PMR (SOCS3), PT-INR and IL-6, which may have a good potential clinical application value.
Collapse
Affiliation(s)
- Ji-Hui Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yuna Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Jing Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Xue-Fei Wei
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Na Wang
- Department of Hepatology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, People's Republic of China
| | - Jing-Wei Wang
- Department of Hepatology, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, People's Republic of China
| | - Hui Lyu
- Department of Severe Liver Disease, Shandong Public Health Clinical Center of Shandong University, Jinan, Shandong, People's Republic of China
| | - Xue-Mei Jiang
- Department of Hepatology, Shandong Public Health Clinical Center of Shandong University, Jinan, Shandong, People's Republic of China
| | - Hui-Hui Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
- Institute of Hepatology, Shandong University, Jinan, Shandong, People's Republic of China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
- Institute of Hepatology, Shandong University, Jinan, Shandong, People's Republic of China
- Shenzhen Research Institute, Shandong University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
3
|
Liu W, Ni Y, Wang W, Cui K, Duan Q, Bai Z, Shahzad A, Liu X, Dong Y, Xu Z, Zhang J, Peng D, Teng Z, Gao Y, Yang Z, Zhang Q. SEPT5 overexpression predicts poor prognosis and promotes progression through feedback regulation of HIF-1α in clear cell renal cell carcinoma. Cell Signal 2025; 127:111591. [PMID: 39798774 DOI: 10.1016/j.cellsig.2025.111591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC), a predominant subtype of renal cell carcinoma, significantly contributes to the heightened morbidity and mortality in individuals diagnosed with urologic tumors. The challenges posed by high malignancy at the initial diagnosis of ccRCC, therapeutic resistance, and unfavorable patient prognosis remain largely unresolved. Our findings indicate that SEPT5 is upregulated in ccRCC and this upregulation is associated with an adverse prognosis for ccRCC patients. Furthermore, we demonstrate that overexpression of SEPT5 promotes proliferation of ccRCC cells, alters their cell cycle distribution, and enhances their migratory and invasive capabilities. Additionally, we observe a positive correlation between SEPT5 overexpression and resistance to sorafenib and sunitinib in ccRCC cells. Further mechanistic investigations have revealed that SEPT5 serves as a novel direct transcriptional target of HIF-1α, leading to subsequent reduction in protein expression and nuclear translocation of HIF-1α. This establishes a feedback loop in ccRCC tumorigenesis. Ultimately, knockdown of SEPT5 significantly inhibits xenografted tumor growth in vivo. Overall, this study provides compelling evidence that directly targeting the HIF-1α-SEPT5 feedback axis may be an effective approach for suppressing the proliferation and progression of ccRCC, offering new insights into the diagnosis and treatment of ccRCC patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/genetics
- Cell Line, Tumor
- Animals
- Prognosis
- Cell Proliferation
- Mice, Nude
- Septins/metabolism
- Septins/genetics
- Male
- Gene Expression Regulation, Neoplastic
- Cell Movement
- Feedback, Physiological
- Disease Progression
- Female
- Mice
- Drug Resistance, Neoplasm/genetics
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
Collapse
Affiliation(s)
- Wenjing Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yueli Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Wenjie Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Kun Cui
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Qiuxin Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Ziyuan Bai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China; Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Asif Shahzad
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Xiangjie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yurong Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China; Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Jinshan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Dongmei Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China; Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Zhuoran Teng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China.
| | - Yanping Gao
- Departments of Radiotherapy, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China.
| | - Zhe Yang
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China.
| |
Collapse
|
4
|
Hajebi Khaniki S, Shokoohi F. Data-Driven Identification of Early Cancer-Associated Genes via Penalized Trans-Dimensional Hidden Markov Models. Biomolecules 2025; 15:294. [PMID: 40001597 PMCID: PMC11853217 DOI: 10.3390/biom15020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is a significant worldwide health problem due to its high prevalence, mortality rates, and frequent diagnosis at advanced stages. While diagnostic and therapeutic approaches have evolved, the underlying mechanisms driving CRC initiation and progression are not yet fully understood. Early detection is critical for improving patient survival, as initial cancer stages often exhibit epigenetic changes-such as DNA methylation-that regulate gene expression and tumor progression. Identifying DNA methylation patterns and key survival-related genes in CRC could thus enhance diagnostic accuracy and extend patient lifespans. In this study, we apply two of our recently developed methods for identifying differential methylation and analyzing survival using a sparse, finite mixture of accelerated failure time regression models, focusing on key genes and pathways in CRC datasets. Our approach outperforms two other leading methods, yielding robust findings and identifying novel differentially methylated cytosines. We found that CRC patient survival time follows a two-component mixture regression model, where genes CDH11, EPB41L3, and DOCK2 are active in the more aggressive form of CRC, whereas TMEM215, PPP1R14A, GPR158, and NAPSB are active in the less aggressive form.
Collapse
Affiliation(s)
- Saeedeh Hajebi Khaniki
- Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad 9137673119, Iran;
| | - Farhad Shokoohi
- Department of Mathematical Sciences, University of Nevada Las Vegas, Las Vegas, NV 89154, USA
| |
Collapse
|
5
|
Jin D, Qian L, Chen J, Yu Z, Dong J. Diagnostic accuracy of methylated SEPT9 for primary liver cancer: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2025; 16:1434174. [PMID: 40017695 PMCID: PMC11864958 DOI: 10.3389/fendo.2025.1434174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Background Primary live cancer (PLC), including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). This meta-analysis was conducted to evaluate the diagnostic efficacy of blood methylated septin 9 gene (mSEPT9) for PLC and to analyze its performance across various subgroups. Methods We conducted a comprehensive search across PubMed, the Cochrane Library, Embase, and China National Knowledge Infrastructure (CNKI), covering research up to May 10, 2024. The pooled sensitivity, specificity, diagnostic odds ratios, and area under the summary receiver operating characteristic (AUC) were calculated for the diagnostic performance of mSEPT9 for PLC. The quality of the studies was assessed using the QUADAS-2 tool, and the meta-analysis was performed using Stata16.0 software. Results Ten articles with 2,182 participants were included in the meta-analysis. The pooled sensitivity of mSEPT9 for detecting primary liver cancer was 0.51 (95% confidence interval [CI]: 0.37-0.65), and the pooled specificity was 0.93 (95% CI: 0.78-0.98). The pooled diagnostic odds ratio was 13 (95% CI: -58), and the area under the Summary Receiver Operator Characteristic Curve was 0.75 (95% CI: 0.71-0.79). Subgroup analyses showed that ICC, case-control studies, qPCR and Asian populations had higher specificities (0.99 [95% CI: 0.97-1.00], 0.93 [95% CI: 0.91-0.95], 0.90 [95% CI: 0.88-0.92] and 0.94 [95% CI: 0.92-0.96], respectively) and diagnostic odds ratios (62.04 [95% CI: 6.53-589.53], 17.62 [95% CI: 4.03-76.99], 13.03 [95% CI: 2.01-84.63] and 14.19 [95% CI: 2.42-83.11], respectively) compared to hepatocellular carcinoma, cohort Study, and Euramerican populations. Conclusions This study confirmed that mSEPT9 in blood has high specificity and moderate sensitivity for detecting primary liver cancer. The diagnostic performance of mSEPT9 varied across different subgroups, limiting its use as an independent screening tool and necessitating its use in conjunction with other methods for confirmatory diagnostics. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024549669.
Collapse
Affiliation(s)
- Danwen Jin
- Pathological Diagnosis Center, Zhoushan Hospital, Zhoushan, Zhejiang, China
| | - Liyong Qian
- Pathological Diagnosis Center, Zhoushan Hospital, Zhoushan, Zhejiang, China
| | - Jiayao Chen
- Department of Laboratory, Zhoushan Hospital, Zhoushan, Zhejiang, China
| | - Ze Yu
- Laboratory of Cell Biology and Molecular Biology, Zhoushan Hospital, Zhoushan, Zhejiang, China
| | - Jinliang Dong
- Department of Hepatobiliary Surgery, Zhoushan Hospital, Zhoushan, Zhejiang, China
| |
Collapse
|
6
|
Schrom EC, McCaffrey EF, Sreejithkumar V, Radtke AJ, Ichise H, Arroyo-Mejias A, Speranza E, Arakkal L, Thakur N, Grant S, Germain RN. Spatial Patterning Analysis of Cellular Ensembles (SPACE) finds complex spatial organization at the cell and tissue levels. Proc Natl Acad Sci U S A 2025; 122:e2412146122. [PMID: 39903116 PMCID: PMC11831171 DOI: 10.1073/pnas.2412146122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/27/2024] [Indexed: 02/06/2025] Open
Abstract
Spatial patterns of cells and other biological elements drive physiologic and pathologic processes within tissues. While many imaging and transcriptomic methods document tissue organization, discerning these patterns is challenging, especially when they involve multiple elements in complex arrangements. To address this challenge, we present Spatial Patterning Analysis of Cellular Ensembles (SPACE), an R package for analysis of high-plex spatial data. SPACE is compatible with any data collection modality that records values (i.e., categorical cell/structure types or quantitative expression levels) at fixed spatial coordinates (i.e., 2d pixels or 3d voxels). SPACE detects not only broad patterns of co-occurrence but also context-dependent associations, quantitative gradients and orientations, and other organizational complexities. Via a robust information theoretic framework, SPACE explores all possible ensembles of tissue elements-single elements, pairs, triplets, and so on-and ranks the most strongly patterned ensembles. For single images, rankings reflect differences from random assortment. For sets of images, rankings reflect differences across sample groups (e.g., genotypes, treatments, timepoints, etc.). Further tools then characterize the nature of each pattern for intuitive interpretation. We validate SPACE and demonstrate its advantages using murine lymph node images for which ground truth has been defined. We then detect new patterns across varied datasets, including tumors and tuberculosis granulomas.
Collapse
Affiliation(s)
- Edward C. Schrom
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Erin F. McCaffrey
- Spatial Immunology Unit, T-Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Vivek Sreejithkumar
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Andrea J. Radtke
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Armando Arroyo-Mejias
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Emily Speranza
- Florida Research and Innovation Center, Cleveland Clinic Lerner Research Institute, Port Saint Lucie, FL34987
| | - Leanne Arakkal
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Nishant Thakur
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| | - Spencer Grant
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, NIH, Bethesda, MD20892-1892
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892-1892
| |
Collapse
|
7
|
Ye M, Xu G, Zhang L, Kong Z, Qiu Z. Meta Analysis of Methylenetetrahydrofolate Reductase (MTHFR) C677T polymorphism and its association with folate and colorectal cancer. BMC Cancer 2025; 25:169. [PMID: 39875876 PMCID: PMC11776141 DOI: 10.1186/s12885-025-13546-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND DNA hypomethylation and uracil misincorporation into DNA, both of which have a very important correlation with colorectal carcinogenesis. Folate plays a crucial role in DNA synthesis, acting as a coenzyme in one-carbon metabolism, which involves the synthesis of purines, pyrimidines, and methyl groups. MTHFR, a key enzyme in folate metabolism, has been widely studied in relation to neural tube defects and hypertension, but its role in colorectal cancer remains underexplored. Therefore, understanding the role of folate and MTHFR genes in colorectal cancer may be helpful for potential preventive or therapeutic interventions. In this meta-analysis, the effects of MTHFR genotype and folate intake on colorectal cancer incidence were analyzed. METHODS We searched PubMed,Embase, Web of Science, and CNKI database to identify relevant studies up to January 2024. We included a series of studies on the association of MTHFR C677T genotype and folate intake with colorectal cancer incidence. The meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols (PRISMA-P). It included 100 studies (39702 cases and 55718 controls),that investigated the association between the MTHFR C677T polymorphism and colorectal cancer (CRC). Additionally, the analysis incorporated further stratification by ethnic population and geographical region. Furthermore, Six of the studies which clarified high amount of folate might be a protective factor for CRC in all three MTHFR C677T genotype, especially in TT genotype. RESULTS MTHFR 677TT genotype was negatively associated with CRC incidence compared with CC genotype (OR = 0.89; 95% CI: 0.85-0.93; P < 0.00001; Z = 5.17). MTHFR 677CT genotype was not significantly associated with colorectal cancer incidence (OR = 1.00; 95% CI: 0.98,1.03). A negative correlation between TT genotype and CRC was observed in ethnics of Asians (OR = 0.83, 95% CI: 0.76, 0.91), Caucasians (OR = 0.93, 95% CI: 0.88, 0.99) and the region of USA (OR = 0.77, 95% CI: 0.71, 0.85), Asia (OR = 0.93, 95% CI: 0.86, 1.00) and Europe (OR = 0.93, 95% CI:0.87, 1.00),but not in Indian (TT: OR = 1.67, 95% CI: 1.06, 2.63; CT: OR = 1.31, 95% CI: 1.00, 1.73)). Amount folate intakes might reduce the morbidity of CRC for people in MTHFR 677TT genotype (OR = 0.68; 95% CI: 0.48,0.96; P = 0.03). CONCLUSION The analysis showed that the incidence of colorectal cancer was reduced among individuals with TT genotype. The individuals with TT genotype and amount folate intake may collectively improve the incidence of colorectal cancer. While the MTHFR 677TT genotype is associated with a reduced risk of CRC, especially in certain populations, these findings should be interpreted with caution due to the limitations of retrospective studies.
Collapse
Affiliation(s)
- Meng Ye
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524023, P.R. China
| | - Guojie Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
| | - Liming Zhang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524023, P.R. China
| | - Zhihui Kong
- Department of Laboratory Medicine, Jingshan People's Hospital, Jingshan, 431800, P.R. China.
| | - Zhenhua Qiu
- Department of Laboratory Medicine, Affiliated Gaozhou People's Hospital, Guangdong Medical University, Maoming, 525200, P.R. China.
| |
Collapse
|
8
|
Müller D, Győrffy B. EpigenPlot: An interactive web platform for DNA methylation-based biomarker and drug target discovery in colorectal cancer. Br J Pharmacol 2025. [PMID: 39871596 DOI: 10.1111/bph.17455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND AND PURPOSE Genome-wide methylation studies have significantly advanced our understanding of colorectal adenocarcinoma progression and biomarker discovery. Aberrant DNA methylation plays a crucial role in gene expression regulation during cancer transformation, highlighting the need to identify differentially methylated regions (DMRs) as potential diagnostic and therapeutic markers. However, an integrated resource to explore and validate methylation alterations across colorectal cancer stages has been lacking. We aimed to develop a platform that integrates existing methylation data, systematically identifies DMRs and provides a tool for further investigation. EXPERIMENTAL APPROACH We created a database combining Illumina HumanMethylation450K and EPIC data from normal colon, adenoma and adenocarcinoma tissues, comprising 2346 samples from 19 datasets. Methylation levels were analysed in six gene regions, and comparisons between tissue types were made using Mann-Whitney, Kruskal-Wallis and ROC tests. KEY RESULTS Both adenoma and adenocarcinoma samples exhibited a general decrease in methylation compared to healthy tissue. Differential methylation in genes such as ITGA4, NPY, IGFL1 and LRRC4 was validated. The strongest DMRs were observed in the C1orf70 gene's 5'UTR and TSS200 regions, with AUC values of 0.98 in both of the HM450K and EPIC datasets. We established an interactive web-based platform accessible at https://epigenplot.com/ enabling future analysis of individual gene regions. CONCLUSIONS AND IMPLICATIONS Our study provides an integrated database of DNA methylation profiles across normal, adenoma and adenocarcinoma tissues, offering a valuable resource for biomarker discovery. The integrated web platform can serve as a tool for the development of methylation-based therapies in the future.
Collapse
Affiliation(s)
- Dalma Müller
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, Pecs, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Hungarian Research Network, Budapest, Hungary
| |
Collapse
|
9
|
Cao L, Duan Q, Zhu Z, Xu X, Liu J, Li B. Liquid biopsy technologies: innovations and future directions in breast cancer biomarker detection. Biomed Microdevices 2025; 27:4. [PMID: 39849252 DOI: 10.1007/s10544-025-00734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 01/25/2025]
Abstract
Globally, breast cancer is the most frequent type of cancer, and its early diagnosis and screening can significantly improve the probability of survival and quality of life of those affected. Liquid biopsy-based targets such as circulating tumor cells, circulating tumor DNA, and exosomes have been instrumental in the early discovery of cancer, and have been found to be effective in stage therapy, recurrence monitoring, and drug selection. Biosensors based on these target related biomarkers convert the tested substances into quantifiable signals such as electrical and optical signals through signal transduction, which has the advantages of high sensitivity, simple operation, and low invasiveness. This review provides an overview of the latest progress of liquid biopsy biomarkers in the diagnosis, prognosis and treatment of breast cancer, compares the application and advantages of different biosensors based on these biomarkers in the diagnosis of breast cancer, and analyzes the limitations and solutions of biosensor based methods.
Collapse
Affiliation(s)
- Linhong Cao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Qingli Duan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Zixin Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Xuejing Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| |
Collapse
|
10
|
Liu Y, Hrit JA, Chomiak AA, Stransky S, Hoffman JR, Tiedemann RL, Wiseman AK, Kariapper LS, Dickson BM, Worden EJ, Fry CJ, Sidoli S, Rothbart SB. DNA hypomethylation promotes UHRF1-and SUV39H1/H2-dependent crosstalk between H3K18ub and H3K9me3 to reinforce heterochromatin states. Mol Cell 2025; 85:394-412.e12. [PMID: 39631394 PMCID: PMC11741932 DOI: 10.1016/j.molcel.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/17/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Mono-ubiquitination of lysine 18 on histone H3 (H3K18ub), catalyzed by UHRF1, is a DNMT1 docking site that facilitates replication-coupled DNA methylation maintenance. Its functions beyond this are unknown. Here, we genomically map simultaneous increases in UHRF1-dependent H3K18ub and SUV39H1/H2-dependent H3K9me3 following DNMT1 inhibition. Mechanistically, transient accumulation of hemi-methylated DNA at CpG islands facilitates UHRF1 recruitment and E3 ligase activity toward H3K18. Notably, H3K18ub enhances SUV39H1/H2 methyltransferase activity and, in colon cancer cells, nucleates new H3K9me3 domains at CpG island promoters of DNA methylation-silenced tumor suppressor genes (TSGs). Disrupting UHRF1 enzyme activity prevents H3K9me3 accumulation while promoting PRC2-dependent H3K27me3 as a tertiary layer of gene repression in these regions. By contrast, disrupting H3K18ub-dependent SUV39H1/H2 activity enhances the transcriptional activating and antiproliferative effects of DNMT1 inhibition. Collectively, these findings reveal roles for UHRF1 and H3K18ub in regulating a hierarchy of repressive histone methylation signaling and rationalize a combination strategy for epigenetic cancer therapy.
Collapse
Affiliation(s)
- Yanqing Liu
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Joel A Hrit
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Alison A Chomiak
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Ashley K Wiseman
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Leena S Kariapper
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Bradley M Dickson
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Evan J Worden
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Scott B Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
11
|
Tian R, Zhang H, Wang C, Zhou S, Zhang L, Wang H. Research on Prediction of Multiple Degenerative Diseases and Biomarker Screening Based on DNA Methylation. Int J Mol Sci 2025; 26:313. [PMID: 39796173 PMCID: PMC11719970 DOI: 10.3390/ijms26010313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The aging process will lead to a gradual functional decline in the human body, and even accelerate a significantly increased risk of degenerative diseases. DNA methylation patterns change markedly with one's age, serving as a biomarker of biological age and closely linked to the occurrence and progression of age-related diseases. Currently, diagnostic methods for individual degenerative diseases are relatively mature. However, aging often accompanies the onset of multiple degenerative diseases, presenting certain limitations in existing diagnostic models. Additionally, some identified DNA methylation biomarkers are typically applicable to only one or a few types of cancer or diseases, further restricting their utility. We endeavor to screen for biomarkers associated with multiple degenerative diseases from the perspective of aging-related co-morbid mechanisms and to perform multiple degenerative disease diagnoses. In this study, we explored research based on methylation correlations and patterns to investigate shared mechanisms across multiple degenerative diseases, identifying a set of biomarkers associated with them. We validated these biomarkers with biological omics analysis and the prediction of multiple classes of degenerative diseases, screened the biomarkers from 600 to 110 by biological omics analysis, and demonstrated the validity and predictive ability of the screened 110 biomarkers. We propose a disease diagnostic model based on a multi-scale one-dimensional convolutional neural network (MSDCNN) and a multi-class degenerative disease prediction model (ResDegNet). The two models are well trained and tested to accurately diagnose diseases and categorize four types of degenerative diseases. The research identified 110 biomarkers associated with degenerative diseases, providing a foundation for further exploration of age-related degenerative conditions. This work aims to facilitate early diagnosis, the identification of biomarkers, and the development of therapeutic targets for drug interventions.
Collapse
Affiliation(s)
- Ruoting Tian
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Hao Zhang
- School of Information Science and Engineering (School of Software), Yanshan University, Qinhuangdao 066000, China;
| | - Chencai Wang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Shengyang Zhou
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Li Zhang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun 130117, China;
| |
Collapse
|
12
|
Qin B, Niu H, Qiu L, Zhou H, Lyu P. Fecal methylated syndecan-2 ( SDC2) testing for early screening of colorectal cancerous and precancerous lesions: A real-world retrospective study in China. CANCER PATHOGENESIS AND THERAPY 2025; 3:60-67. [PMID: 39872370 PMCID: PMC11764036 DOI: 10.1016/j.cpt.2024.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 01/30/2025]
Abstract
Background Colorectal cancer (CRC) is a major public health concern and the second leading cause of cancer-related deaths worldwide. However, challenges remain in deploying effective screening strategies for early-stage CRC. This study aimed to evaluate the effectiveness of a fecal-based syndecan-2 (SDC2) methylation test for the detection of colorectal lesions and CRC. Methods We retrospectively collected data on participants who underwent fecal SDC2 methylation testing from January 1, 2019, to May 30, 2023. Patients with positive results were recommended to undergo colonoscopy. Performance indicators associated with certain clinical characteristics, including positive rate (PR), positive predictive value (PPV), and colonoscopy compliance rate (CCR), were subjected to statistical analysis. Results We analyzed data from 113,209 participants, of whom 11,841 (10.4% PR) had positive fecal SDC2 methylation test results. A total of 4315 participants with positive results adhered to the colonoscopy recommendations, and the CCR was 36.4%. Finally, 3169 colorectal lesions were detected, including 1134 polyps, 875 non-advanced adenomas (NAAs), 770 advanced adenomas (AAs), and 390 CRCs, with PPV values of 26.3% (1134/4315), 20.3% (875/4315), 17.8% (770/4315), and 9.0% (390/4315), respectively. Notably, the PPV for CRC increased significantly with age (χ 2 = 164.40, P < 0.0001). In addition, as the cycle threshold (CT) values increased, the PPVs of AAs and CRCs generally decreased, whereas those of NAAs and polyps significantly increased. Moreover, the clinical patient group had the highest incidence of late-stage CRC (stage II and higher), whereas asymptomatic populations from the staff physical examination group and rural town-based screening programs had the highest number of stage 0 and I CRCs detected (P = 0.0107). Conclusions This study indicates that fecal SDC2 methylation testing combined with colonoscopy may be an effective screening method for colorectal lesions and CRC.
Collapse
Affiliation(s)
- Boyu Qin
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Haitao Niu
- National Health Commission of the People's Republic of China, Beijing 100044, China
| | - Lupeng Qiu
- Medical School of Chinese PLA, Beijing 100853, China
| | - Hongfeng Zhou
- Health Management Department, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong 510000, China
| | - Peng Lyu
- Cancer Pathogenesis and Therapy, Chinese Medical Association Publishing House, Beijing 100052, China
- Key Laboratory of Knowledge Mining and Service for Medical Journals, National Press and Publication Administration, Beijing 100052, China
- Beijing Beiya Hospital of Traditional Chinese Medicine, Beijing 100029, China
| |
Collapse
|
13
|
Zhang W, Jie W, Cui W, Duan G, Zou Y, Peng X. DMRIntTk: Integrating different DMR sets based on density peak clustering. PLoS One 2024; 19:e0315920. [PMID: 39715163 DOI: 10.1371/journal.pone.0315920] [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: 08/03/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Identifying differentially methylated regions (DMRs) is a basic task in DNA methylation analysis. However, due to the different strategies adopted, different DMR sets will be predicted on the same dataset, which poses a challenge in selecting a reliable and comprehensive DMR set for downstream analysis. RESULTS Here, we develop DMRIntTk, a toolkit for integrating DMR sets predicted by different methods on a same dataset. In DMRIntTk, the genome is segmented into bins, and the reliability of each DMR set at different methylation thresholds is evaluated. Then, the bins are weighted based on the covered DMR sets and integrated into final DMRs using a density peak clustering algorithm. To demonstrate the practicality of DMRIntTk, it was applied to different scenarios, including tissues with relatively large methylation differences, cancer tissues versus normal tissues with medium methylation differences, and disease tissues versus normal tissues with subtle methylation differences. Our results show that DMRIntTk can effectively trim regions with small methylation differences from the original DMR sets and thereby enriching the proportion of DMRs with larger methylation differences. In addition, the overlap analysis suggests that the integrated DMR sets are quite comprehensive, and functional analyses indicate the integrated disease-related DMRs are significantly enriched in biological pathways associated with the pathological mechanisms of the diseases. A comparative analysis of the integrated DMR set versus each original DMR set further highlights the superiority of DMRIntTk, demonstrating the unique biological insights it can provide. CONCLUSIONS Conclusively, DMRIntTk can help researchers obtain a reliable and comprehensive DMR set from many prediction methods.
Collapse
Affiliation(s)
- Wenjin Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Wenlong Jie
- Hunan Key Laboratory of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
| | - Wanxin Cui
- Hunan Key Laboratory of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
| | - Guihua Duan
- Hunan Key Laboratory of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
| | - You Zou
- Hunan Key Laboratory of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, China
- High Performance Computing Center, Central South University, Changsha, China
| | - Xiaoqing Peng
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
14
|
Lyu G, Sun R, Liu X, Xu Z. A Novel Hypoxia-Featured Genes Prognostic Model for Identification of Hypoxia Subtypes in Diffuse Large B-Cell Lymphoma. Cell Biochem Biophys 2024:10.1007/s12013-024-01637-7. [PMID: 39663278 DOI: 10.1007/s12013-024-01637-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL), known as the predominant type of aggressive B-cell lymphoma, is biologically and clinically heterogeneous. The prognosis of DLBCL is quite different among subtypes. Hypoxia is one of the key elements in tumor microenvironment, promoting tumor progression by means of various mechanisms, such as increased proliferation, altered metabolism, enhanced angiogenesis, and greater migratory capability, among others. The primary purpose of this research is to investigate the connection between hypoxia-featured genes (HFGs), prognosis in DLBCL, and their capacity association with the immune microenvironment. Various hypoxia-associated patterns for DLBCL patients from GEO and TCGA databases were identified by means of an unsupervised consensus clustering algorithm. CIBERSORT and IOBR package is used to identify different immune infiltration status. To develop a predictive model using hypoxia-related genes, we conducted univariate Cox regression, multivariate Cox regression, and LASSO regression assessment. Subsequently, we confirmed the predictive importance of these hypoxia-associated genes, highlighting hypoxia-associated characteristics, and explored the connection between the hypoxia model and the immune environment. Three hypoxia clusters were identified. We also observed that each pattern of hypoxia response was significantly related to different prognoses. It was found that the immune status among hypoxia clusters is different. After developing a prognostic risk model using 5 hypoxia-related genes, we discovered that the risk score is related to immune factors and how effective drugs are in treating DLBCL. In DLBCL patients, varying hypoxia patterns correlate with both prognostic outcomes and the immune microenvironment. Hypoxia-featured genes (HFGs) function as a standalone predictive element in these patients. It is also potentially a reliable indicator for predicting clinical responses to ICI therapy and traditional drugs.
Collapse
Affiliation(s)
- Geng Lyu
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruixin Sun
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaxin Liu
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zizhen Xu
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
15
|
Hu B, Zhang Y, Jiang B, Li A. Prognostic value of circulating long non-coding RNAs in colorectal cancer patients: a meta-analysis. Expert Rev Anticancer Ther 2024; 24:1249-1259. [PMID: 37934874 DOI: 10.1080/14737140.2023.2280643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
OBJECTIVES This meta-analysis aimed to evaluate the prognostic significance of circulating long non-coding RNAs (lncRNAs) in colorectal cancer (CRC). METHODS A comprehensive literature search was conducted in databases (Embase, Web of Science, PubMed, and Cochrane Library) up to July 2022. The quality of included studies was assessed using the Newcastle-Ottawa Scale (NOS). Statistical analysis was performed with Review Manager 5.4 and Stata 17.0. Publication bias was assessed using Begg's test, and sensitivity analysis was conducted to validate the meta-analysis results. RESULTS Ten articles, comprising 1,473 CRC patients and 18 different circulating lncRNAs, were included. Thirteen circulating lncRNAs were found to be up-regulated in CRC patients, while five were down-regulated. High expression of circulating lncRNAs up-regulated in CRC patients was associated with shorter CRC OS (HR = 2.91, 95% CI: 1.17, 7.22; P = 0.02, I2 = 86%). Conversely, high expression of circulating lncRNAs down-regulated in CRC patients was linked to longer CRC OS (HR = 0.16, 95% CI: 0.07, 0.40; P < 0.0001, I2 = 0%) and improved DFS (HR = 0.52, 95% CI: 0.37, 0.74; P = 0.0002, I2 = 0%). Additionally, circulating lncRNA levels correlated with TNM staging, tumor location, and lymph node metastasis. CONCLUSION Circulating lncRNAs show promise as prognostic markers for CRC patients, but further studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Bin Hu
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, China
| | - Yanfei Zhang
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, China
| | - Bingjing Jiang
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, China
| | - Angcheng Li
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, China
| |
Collapse
|
16
|
Bai J, Wang Z, Yang M, Xiang J, Liu Z. Disrupting CENP-N mediated SEPT9 methylation as a strategy to inhibit aerobic glycolysis and liver metastasis in colorectal cancer. Clin Exp Metastasis 2024; 41:971-988. [PMID: 39424682 DOI: 10.1007/s10585-024-10316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Colorectal cancer (CRC) is a prevalent malignancy with a high mortality rate, primarily due to liver metastasis. This study explores the role of centromere protein N (CENP-N) in mediating the methylation of septin 9 (SEPT9) and its subsequent effects on aerobic glycolysis and liver metastasis in CRC. We employed in vitro and in vivo experiments, including single-cell RNA sequencing, methylation-specific PCR (MSP), ChIP assays, and various functional assays to assess the impact of CENP-N and SEPT9 on CRC cell proliferation, migration, invasion, and metabolic reprogramming. Our data reveal that CENP-N directly interacts with SEPT9, enhancing its methylation at specific lysine residues. This modification significantly upregulates key glycolytic enzymes, thereby promoting aerobic glycolysis, CRC cell proliferation, and migration. In vivo studies further demonstrate that the CENP-N/SEPT9 axis facilitates liver metastasis of CRC, as confirmed by fluorescence imaging and histological analysis. This study identifies a novel pathway where CENP-N-mediated methylation of SEPT9 drives metabolic reprogramming and metastasis in CRC. These findings suggest potential therapeutic targets for inhibiting CRC progression and liver metastasis, offering new insights into CRC pathogenesis.
Collapse
Affiliation(s)
- Junge Bai
- Department of Biochemistry and Molecular Biology, Harbin Medical University, 157 Health Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Zhexue Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Ming Yang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Jun Xiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
17
|
Xu W, Li W, Li Y, Kuai D, Sun W, Liu X, Xu B. Genetic insights into blood protein correlations with colorectal cancer: a Mendelian randomization study. Discov Oncol 2024; 15:710. [PMID: 39586851 PMCID: PMC11589031 DOI: 10.1007/s12672-024-01584-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Several blood proteins might be associated with the development of colorectal cancer (CRC), but many studies on this topic are often biased. By using genetic variation data, which is less influenced by environmental factors, we can better determine the causal relationship between specific blood proteins and the occurrence of colorectal cancer. METHODS Data from a genome-wide association study (GWAS) on blood proteins, encompassing 1,478 proteins, and colorectal cancer (CRC) GWAS data, covering 637,693 subjects, were collected and organized. Additionally, GWAS data for obesity, diabetes mellitus (DM), and smoking were obtained for further analysis. Single nucleotide polymorphisms (SNPs) significantly associated with the exposure factors (blood proteins) were selected to ensure their independence from other confounding factors and outcomes (CRC onset), and that they only affected outcomes through blood proteins. The causal effects of Mendelian Randomization (MR) were primarily estimated using the inverse variance weighted (IVW) method, with other methods serving as supplementary approaches. The Cochran's Q-test assessed heterogeneity among SNP estimates; the MR Egger method evaluated pleiotropy; and the leave-one-out test examined the sensitivity of individual SNPs. Obesity, DM, and smoking were included in the multivariate MR analysis. RESULT A total of 31 SNPs and 8 blood protein exposure factors were identified, specifically TNFRSF16 (4 SNPs), RNF8 (4 SNPs), MRM3 (4 SNPs), ST6GALNAC1 (4 SNPs), TIE1 (4 SNPs), CBP (4 SNPs), DNAJB9 (4 SNPs), and EDN2 (3 SNPs). The IVW results showed a significant causal relationship between all 8 exposure factors and colorectal cancer (P < 0.05). Among these, TNFRSF16, TIE1, and EDN2 were identified as risk factors, while the remaining five served as protective factors. The causal inference in this study was not influenced by pleiotropy or environmental factors such as obesity, diabetes, and smoking. Therefore, the results were both stable and reliable. CONCLUSION Eight blood proteins (or genes) have been identified as having a causal relationship with the onset of colorectal cancer, suggesting their potential use as screening biomarkers and treatment targets.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Wei Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Yaqiang Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Dayu Kuai
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Wei Sun
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Xian Liu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China
| | - Baohong Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, 100000, China.
| |
Collapse
|
18
|
El-Gammal MA, Sayed FE, Allam NK. Comprehensive analysis of electrochemical biosensors for early ovarian cancer detection. RSC Adv 2024; 14:37580-37597. [PMID: 39588243 PMCID: PMC11587864 DOI: 10.1039/d4ra05972g] [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: 08/17/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
Ovarian cancer is one of the leading causes of mortality among women worldwide. However, early detection can significantly reduce mortality rates and mitigate subsequent complications related to both economic burden and mental well-being. Despite the development in the field of medical diagnosis, the death rates due to ovarian cancer have sharply increased. Among the recent technologies suggested as suitable diagnostic techniques for the early detection of ovarian cancer, biosensor technology has emerged as a cutting-edge technology, with electrochemical biosensors providing one of the most efficient types of biosensors. Therefore, this review discusses the application of electrochemical biosensors as a viable alternative to conventional diagnostic techniques for the timely identification of ovarian cancer, its advantages over other types of biosensors and conventional diagnostic techniques, and the types of electrochemical biosensors.
Collapse
Affiliation(s)
- Marwa A El-Gammal
- Energy Materials Laboratory, Physics Department, School of Sciences and Enginnering, The American University in Cairo New Cairo 11835 Egypt
| | - Fatma E Sayed
- Biotechnology program, Faculty of Agriculture, Ain Shams University Cairo 11566 Egypt
| | - Nageh K Allam
- Energy Materials Laboratory, Physics Department, School of Sciences and Enginnering, The American University in Cairo New Cairo 11835 Egypt
| |
Collapse
|
19
|
Zhang L, Wang D, Li Z, Lin G, Li J, Zhang R. Interlaboratory consistency of SDC2 promoter methylation detection in colorectal cancer using the post-optimized materials. iScience 2024; 27:111177. [PMID: 39569371 PMCID: PMC11577190 DOI: 10.1016/j.isci.2024.111177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/05/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024] Open
Abstract
Fecal DNA-based Syndecan 2 (SDC2) methylation detection is a promising non-invasive strategy for early colorectal cancer (CRC) screening. In China, commercial assays for SDC2 methylation detection vary in sensitivity and specificity, yet there is no standardized external quality assessment (EQA) to ensure accuracy. This study utilized CRISPR-Cas9 and homology-directed repair (HDR) technologies to edit the SDC2 promoter in 293T cells, creating hypermethylated and heterogeneous cell lines. These cell lines were used to develop an EQA panel for SDC2 methylation. We established a 10-sample panel, encompassing a range of methylation levels, and conducted an EQA across 140 laboratories. Among 1,400 results, 0.57% were incorrect. The optimized EQA materials effectively monitor the accuracy of SDC2 methylation detection in CRC, supporting reliable and consistent clinical testing and contributing to early CRC screening and diagnosis in China.
Collapse
Affiliation(s)
- Lijing Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Duo Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Ziqiang Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Guigao Lin
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Jinming Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Rui Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| |
Collapse
|
20
|
Yang K, Yang Y, Yu L, Yang F, Xiang Y, Zeng J, Huang N. Methylation modification is a poor prognostic factor in non-small cell lung Cancer and regulates the tumor microenvironment: mRNA molecular structure and function. Int J Biol Macromol 2024; 282:137214. [PMID: 39491713 DOI: 10.1016/j.ijbiomac.2024.137214] [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: 09/04/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the most common and deadly type of lung cancer, and its poor prognosis is closely related to the complex interactions of the tumor microenvironment. Through methylation analysis of tumor tissue samples from NSCLC patients, combined with high-throughput sequencing technology, the methylation status and structural characteristics of mRNA molecules were studied. Bioinformatics tools were used to analyze the regulatory effects of methylation modification on mRNA expression and genes associated with the tumor microenvironment. The results showed that the methylation level of specific mRNA was significantly correlated with the expression changes of tumor microenvironment-related factors. In addition, methylation modification affected mRNA stability and translation efficiency, further altering the metabolic activity and immune escape capacity of tumor cells. The results showed that mRNA with high methylation level was significantly associated with poor prognosis. Methylation modification profoundly affects the tumor microenvironment and prognosis of non-small cell lung cancer by altering the structure and function of mRNA molecules.
Collapse
Affiliation(s)
- Kai Yang
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - YuPing Yang
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Lin Yu
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fan Yang
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - YuXin Xiang
- School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jun Zeng
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
| | - Na Huang
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institute, Chengdu, Sichuan, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China; School Of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
21
|
Yun P, Kulaixijiang K, Pan J, Yang L, Wang N, Xu Z, Zhang Y, Cai H, Zhao ZY, Zhu M, Yan H. Early colorectal cancer diagnosis: A novel methylated stool DNA model enhanced the diagnostic efficiency. United European Gastroenterol J 2024. [PMID: 39487820 DOI: 10.1002/ueg2.12696] [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: 04/27/2024] [Accepted: 08/18/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Methylated stool DNA (sDNA) is a reliable noninvasive biomarker for early colorectal cancer (CRC) diagnosis. However, there are barely any diagnostic panels that can achieve both a sensitivity and specificity exceeding 90% simultaneously. OBJECTIVE We aimed to identify a novel methylated sDNA panel and model for the early diagnosis of CRC. METHODS We conducted methyl-CpG binding domain isolated genome sequencing (MiGS) on CpG island methylation phenotype (CIMP)-positive (n = 3) and CIMP-negative CRC tissues (n = 3) and their corresponding normal adjacent tissues. Subsequently, by utilizing both the aforementioned data and public datasets, we identified a set of promising methylated sDNA markers for CRC. Next, we validated 5 of these genes using pyrosequencing in CRC patients (n = 31). Then, we developed a combined diagnostic model (CDM) for CRC based on the methylation status of PRDM12, FOXE1, and SDC2 by a Training cohort (n = 231). Finally, the performance of CDM was evaluated in an independent multicenter Validation cohort (n = 800). RESULTS A total of 1062 participants were included in this study. The area under the curve (AUC) of the CDM was 0.979 (95% CI: 0.960-0.997), and the optimal sensitivity and specificity were 97.35% and 99.05%, respectively, in the training cohort (n = 231). In the independent validation cohort (n = 800), the AUC was 0.950 (95% CI: 0.927-0.973), along with the optimal sensitivity of 92.75% and specificity of 97.21%. When CRC and advanced adenoma (AAD) were used as diagnostic targets, the model AUC was 0.945 (95% CI: 0.922-0.969), with an optimal sensitivity of 91.89% and a specificity of 95.21%. The model sensitivity for nonadvanced adenoma patients was 68.66%. CONCLUSION The sDNA diagnostic model CDM, developed from both CIMP-P and CIMP-N, exhibited exceptional performance in CRC and could serve as a potential alternative strategy for CRC screening.
Collapse
Affiliation(s)
- Peng Yun
- Reproductive Medicine Center, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Kamila Kulaixijiang
- Department of Pathology, Karamay Central Hospital of Xinjiang, Karamay, China
- Xinjiang Key Laboratory of Clinical Genetic Testing and Biomedical Information, Karamay, China
- Xinjiang Clinical Research Center for Precision Medicine of Digestive System Tumors, Karamay, China
| | - Jiang Pan
- Reproductive Medicine Center, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Luping Yang
- Reagent R&D Department, Xiamen Sciendox Biotechnology Co., Ltd., Xiamen, China
| | - Nengzhuang Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zheng Xu
- Central Laboratory, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yaodong Zhang
- Reproductive Medicine Center, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Clinical Lab, The 971st PLA Navy Hospital, Qingdao, China
| | - Haifang Cai
- Reagent R&D Department, Xiamen Sciendox Biotechnology Co., Ltd., Xiamen, China
| | - Zi-Ye Zhao
- Department of Colorectal Surgery and Hereditary Colorectal Cancer Registry, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Min Zhu
- Department of Pathology, Karamay Central Hospital of Xinjiang, Karamay, China
- Xinjiang Key Laboratory of Clinical Genetic Testing and Biomedical Information, Karamay, China
- Xinjiang Clinical Research Center for Precision Medicine of Digestive System Tumors, Karamay, China
| | - Hongli Yan
- Reproductive Medicine Center, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
22
|
Luo S, Yue M, Wang D, Lu Y, Wu Q, Jiang J. Breaking the barrier: Epigenetic strategies to combat platinum resistance in colorectal cancer. Drug Resist Updat 2024; 77:101152. [PMID: 39369466 DOI: 10.1016/j.drup.2024.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Platinum-based drugs, such as cisplatin and oxaliplatin, are frontline chemotherapy for CRC, effective in both monotherapy and combination regimens. However, the clinical efficacy of these treatments is often undermined by the development of drug resistance, a significant obstacle in cancer therapy. In recent years, epigenetic alterations have been recognized as key players in the acquisition of resistance to platinum drugs. Targeting these dysregulated epigenetic mechanisms with small molecules represents a promising therapeutic strategy. This review explores the complex relationship between epigenetic changes and platinum resistance in CRC, highlighting current epigenetic therapies and their effectiveness in countering resistance mechanisms. By elucidating the epigenetic underpinnings of platinum resistance, this review aims to contribute to ongoing efforts to improve treatment outcomes for CRC patients.
Collapse
Affiliation(s)
- Shiwen Luo
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ming Yue
- Department of Pharmacy, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Dequan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yukang Lu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jue Jiang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
23
|
Overs A, Peixoto P, Hervouet E, Molimard C, Monnien F, Durand J, Guittaut M, Vienot A, Viot J, Herfs M, Borg C, Feugeas JP, Selmani Z. COL25A1 and METAP1D DNA methylation are promising liquid biopsy epigenetic biomarkers of colorectal cancer using digital PCR. Clin Epigenetics 2024; 16:146. [PMID: 39425144 PMCID: PMC11490026 DOI: 10.1186/s13148-024-01748-1] [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: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Colorectal cancer is a public health issue and was the third leading cause of cancer-related death worldwide in 2022. Early diagnosis can improve prognosis, making screening a central part of colorectal cancer management. Blood-based screening, diagnosis and follow-up of colorectal cancer patients are possible with the study of cell-free circulating tumor DNA. This study aimed to identify novel DNA methylation biomarkers of colorectal cancer that can be used for the follow-up of patients with colorectal cancer. METHODS A DNA methylation profile was established in the Gene Expression Omnibus (GEO) database (n = 507) using bioinformatics analysis and subsequently confirmed using The Cancer Genome Atlas (TCGA) database (n = 348). The in silico profile was then validated on local tissue and cell-free DNA samples using methylation-specific digital PCR in colorectal cancer patients (n = 35) and healthy donors (n = 35). RESULTS The DNA methylation of COL25A1 and METAP1D was predicted to be a colorectal cancer biomarker by bioinformatics analysis (ROC AUC = 1, 95% CI [0.999-1]). The two biomarkers were confirmed with tissue samples, and the combination of COL25A1 and METAP1D yielded 49% sensitivity and 100% specificity for cell-free DNA. CONCLUSION Bioinformatics analysis of public databases revealed COL25A1 and METAP1D DNA methylation as clinically applicable liquid biopsies DNA methylation biomarkers. The specificity implies an excellent positive predictive value for follow-up, and the high sensitivity and relative noninvasiveness of a blood-based test make these biomarkers compatible with colorectal cancer screening. However, the clinical impact of these biomarkers in colorectal cancer screening and follow-up needs to be established in further prospective studies.
Collapse
Affiliation(s)
- Alexis Overs
- Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France.
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France.
| | - Paul Peixoto
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| | - Eric Hervouet
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| | - Chloé Molimard
- Department of Pathology, University Hospital of Besançon, 25000, Besancon, France
| | - Franck Monnien
- Department of Pathology, University Hospital of Besançon, 25000, Besancon, France
| | - Jules Durand
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| | - Michael Guittaut
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| | - Angélique Vienot
- Department of Oncology, University Hospital of Besançon, 25000, Besancon, France
| | - Julien Viot
- Department of Oncology, University Hospital of Besançon, 25000, Besancon, France
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, Liège, Belgium
| | - Christophe Borg
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
- Department of Oncology, University Hospital of Besançon, 25000, Besancon, France
| | - Jean-Paul Feugeas
- Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| | - Zohair Selmani
- Department of Oncobiology, University Hospital of Besançon, 3 Boulevard Alexandre Fleming, 25000, Besançon, France
- UMR1098, INSERM, University of Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
24
|
Yuan M, Long Q, Sun X. OCTA-based research on changes of retinal microcirculation in digestive tract malignancy. Photodiagnosis Photodyn Ther 2024; 49:104270. [PMID: 39002834 DOI: 10.1016/j.pdpdt.2024.104270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE This cross-sectional study measured retinal vessel density (VD) in patients with digestive tract malignancy by optical coherence tomography angiography (OCTA), and compared them with healthy controls to explore the retinal microcirculation changes in patients with digestive tract malignancy. METHODS 106 eligible participants were divided into three groups: gastric cancer (GC) group (36 individuals), colorectal cancer (CRC) group (34 individuals), and healthy control group (36 individuals). Angio 6 × 6 512 × 512 R4 and ONH Angio 6 × 6 512 × 512 R4 modes were performed to collect retinal vessel density data centered on fovea and papillary, respectively. The retina was automatically segmented into different layers (superficial vascular plexus (SVP), the inner retinal layer, radial peripapillary capillary plexus (RPCP), deep vascular plexus (DVP)) and areas to analyze. RESULTS At the optic nerve head (ONH) region, the VD of the inner retinal layer increased in both GC and CRC groups in all quadrants and areas. In the papillary area, VD in the inner retinal layer, SVP, and RPCP increased in the GC and CRC groups. In the parapapillary area, VD in the inner retinal layer increased in the GC and the CRC groups. Significant increase in the global VD were found in the GC group of the RPCP and SVP. Regarding the macular region, no statistical differences were observed in each layer. CONCLUSIONS The study suggested that retinal vessel density changed in patients with digestive tract malignancy, especially in the inner retinal layer of the ONH region, revealing the potential relevance of the relation between gastrointestinal cancer and retinal microcirculation.
Collapse
Affiliation(s)
- Mingzhu Yuan
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qi Long
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
25
|
Sun J, Shi S, Sun C, Wang J, Yang X, Yang Z, Xu J, Zhang S. Predictive nomogram of the clinical outcomes of colorectal cancer based on methylated SEPT9 and intratumoral IL-10 + Tregs infiltration. J Transl Med 2024; 22:861. [PMID: 39334238 PMCID: PMC11430755 DOI: 10.1186/s12967-024-05635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Gene methylation and the immune-related tumor microenvironment (TME) are highly correlated in tumor progression and therapeutic efficacy. Although both of them can be used to predict the clinical outcomes of colorectal cancer (CRC) patients, their predictive value is still unsatisfactory. Whether a combination risk model comprising these two prediction parameters performs better predictive effectiveness than independent factor is still unclear. Methylated Septin9 (mSEPT9) is an early diagnosis biomarker of CRC, in this study, we aimed to investigate mSEPT9-related biomarkers of immunosuppressive TME and identify the value of the combination risk model in predicting the clinical outcomes of CRC. METHODS Immunofluorescence staining was performed to clarify the correlation between intratumoral IL-10+ Treg infiltration and mSEPT9 in peripheral blood. Survival time, response to 5-fluorouracil (5-FU)-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis were analyzed in study (197 CRC samples) and validation (195 CRC samples) sets to evaluate the efficacy of combination risk model. Potential mechanisms were explored by mRNA sequencing. RESULTS Hypermethylated SEPT9 in the peripheral blood of patients with CRC (stage I-III, and stage IV with resectable M1) before radical resection was positively correlated with high intratumoral IL-10+ Treg infiltration. The high-risk model revealed poor overall survival and progression-free survival, inferior therapeutic response to 5-FU-based chemotherapy and PD-1 blockade, and high probability of recurrence or metastasis. The underlying mechanisms may be associated with the increase in mSEPT9 and mediation of IL-10 via methionine metabolic reprogramming-induced infiltration of IL-10+ Tregs in the TME, which promotes tumor progression and resistance to 5-FU-based chemotherapy and PD-1 blockade. CONCLUSIONS The combination risk model of peripheral mSETP9 and intratumoral IL-10+ Treg infiltration in CRC can effectively predict prognosis, responsiveness to 5-FU-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis. Therefore, this model can be used for precision treatment of CRC.
Collapse
Affiliation(s)
- Jie Sun
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Songli Shi
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300121, People's Republic of China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Zhengduo Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jing Xu
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Department of General Surgery, Tianjin University Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China.
| |
Collapse
|
26
|
Dong L, Li Y, Song X, Sun C, Song X. SFRP1 mediates cancer-associated fibroblasts to suppress cancer cell proliferation and migration in head and neck squamous cell carcinoma. BMC Cancer 2024; 24:1165. [PMID: 39300373 PMCID: PMC11411997 DOI: 10.1186/s12885-024-12907-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), as key cell populations in the tumor microenvironment (TME), play a crucial role in tumor regulation. Previous studies on a prognostic signature of 8 CAF-related genes in head and neck squamous cell carcinoma (HNSCC) revealed that Secreted frizzled-related protein 1 (SFRP1) is one of the hub genes closely related to CAFs. SFRP1 is deficiently expressed in numerous types of cancer and is classified as a tumor suppressor gene. However, the role of SFRP1 in TME regulation in HNSCC remains unclear. This study aimed to explore the role of SFRP1 in the proliferation and migration of HNSCC cells by mediating CAFs and their regulatory mechanisms. METHODS The expression differences, prognosis, and immune infiltration of SFRP1 in HNSCC were analyzed using the TIMER and GEPIA2 databases. The expression of SFRP1 in HNSCC tumor tissues, as well as the expression and secretion of SFRP1 in CAFs and tumor cells, were examined. An indirect co-culture system was constructed to detect the proliferation, migration, and apoptosis of HNSCC cells, and to clarify the effect of SFRP1 on tumor cells by mediating CAFs. Furthermore, the expression and secretion of 10 cytokines derived from CAFs that act on immune cells were verified. RESULTS SFRP1 was differently expressed in HNSCC tumor tissues and highly expressed in CAFs. SFRP1 inhibited the proliferation and migration of tumor cells and promoted apoptosis by mediating CAFs. The detection of CAFs-derived factors suggested that the mechanism of action of SFRP1 was associated with the regulation of immune cells. CONCLUSION SFRP1 inhibits the proliferation and migration of HNSCC cells by mediating CAFs, and the mechanism of action is related to the regulation of immune cells, which may provide new research directions and therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Lei Dong
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Yumei Li
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiaoyu Song
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Qingdao University, Qingdao, China
| | - Caiyu Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| | - Xicheng Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
| |
Collapse
|
27
|
Fan Z, Edelmann D, Yuan T, Köhler BC, Hoffmeister M, Brenner H. Developing survival prediction models in colorectal cancer using epigenome-wide DNA methylation data from whole blood. NPJ Precis Oncol 2024; 8:191. [PMID: 39237753 PMCID: PMC11377733 DOI: 10.1038/s41698-024-00689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
While genome-wide association studies are valuable in identifying CRC survival predictors, the benefit of adding blood DNA methylation (blood-DNAm) to clinical features, including the TNM system, remains unclear. In a multi-site population-based patient cohort study of 2116 CRC patients with baseline blood-DNAm, we analyzed survival predictions using eXtreme Gradient Boosting with a 5-fold nested leave-sites-out cross-validation across four groups: traditional and comprehensive clinical features, blood-DNAm, and their combination. Model performance was assessed using time-dependent ROC curves and calibrations. During a median follow-up of 10.3 years, 1166 patients died. Although blood-DNAm-based predictive signatures achieved moderate performances, predictive signatures based on clinical features outperformed blood-DNAm signatures. The inclusion of blood-DNAm did not improve survival prediction over clinical features. M1 stage, age at blood collection, and N2 stage were the top contributors. Despite some prognostic value, incorporating blood DNA methylation did not enhance survival prediction of CRC patients beyond clinical features.
Collapse
Affiliation(s)
- Ziwen Fan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bruno Christian Köhler
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- NCT Heidelberg, National Center for Tumor Diseases (NCT) a partnership between DKFZ and University Hospital, Heidelberg, Germany.
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
28
|
Yu X, Shao Y, Dong H, Zhang X, Ye G. Biological function and potential application of PANoptosis-related genes in colorectal carcinogenesis. Sci Rep 2024; 14:20672. [PMID: 39237645 PMCID: PMC11377449 DOI: 10.1038/s41598-024-71625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
PANoptosis induces programmed cell death (PCD) through extensive crosstalk and is associated with development of cancer. However, the functional mechanisms, clinical significance, and potential applications of PANoptosis-related genes (PRGs) in colorectal cancer (CRC) have not been fully elucidated. Functional enrichment of key PRGs was analyzed based on databases, and relationships between key PRGs and the immune microenvironment, immune cell infiltration, chemotherapy drug sensitivity, tumor progression genes, single-cell cellular subgroups, signal transduction pathways, transcription factor regulation, and miRNA regulatory networks were systematically explored. This study identified 5 key PRGs associated with CRC: BCL10, CDKN2A, DAPK1, PYGM and TIMP1. Then, RT-PCR was used to verify expression of these genes in CRC cells and tissues. Clinical significance and prognostic value of key genes were further verified by multiple datasets. Analyses of the immune microenvironment, immune cell infiltration, chemotherapy drug sensitivity, tumor progression genes, single-cell cellular subgroups, and signal transduction pathways suggest a close relationship between these key genes and development of CRC. In addition, a novel prognostic nomogram model for CRC was successfully constructed by combining important clinical indicators and the key genes. In conclusion, our findings offer new insights for understanding the pathogenesis of CRC, predicting CRC prognosis, and identifying multiple therapeutic targets for future CRC therapy.
Collapse
Affiliation(s)
- Xuan Yu
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yongfu Shao
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Haotian Dong
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xinjun Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China.
| | - Guoliang Ye
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, China.
- Institute of Digestive Disease of Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
29
|
Kiri S, Ryba T. Cancer, metastasis, and the epigenome. Mol Cancer 2024; 23:154. [PMID: 39095874 PMCID: PMC11295362 DOI: 10.1186/s12943-024-02069-w] [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: 04/30/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Cancer is the second leading cause of death worldwide and disease burden is expected to increase globally throughout the next several decades, with the majority of cancer-related deaths occurring in metastatic disease. Cancers exhibit known hallmarks that endow them with increased survival and proliferative capacities, frequently as a result of de-stabilizing mutations. However, the genomic features that resolve metastatic clones from primary tumors are not yet well-characterized, as no mutational landscape has been identified as predictive of metastasis. Further, many cancers exhibit no known mutation signature. This suggests a larger role for non-mutational genome re-organization in promoting cancer evolution and dissemination. In this review, we highlight current critical needs for understanding cell state transitions and clonal selection advantages for metastatic cancer cells. We examine links between epigenetic states, genome structure, and misregulation of tumor suppressors and oncogenes, and discuss how recent technologies for understanding domain-scale regulation have been leveraged for a more complete picture of oncogenic and metastatic potential.
Collapse
Affiliation(s)
- Saurav Kiri
- College of Medicine, University of Central Florida, 6850 Lake Nona Blvd., Orlando, 32827, Florida, USA.
| | - Tyrone Ryba
- Department of Natural Sciences, New College of Florida, 5800 Bay Shore Rd., Sarasota, 34243, Florida, USA.
| |
Collapse
|
30
|
Jasmine F, Almazan A, Khamkevych Y, Bissonnette M, Ahsan H, Kibriya MG. Association of KRAS Mutation and Gene Pathways in Colorectal Carcinoma: A Transcriptome- and Methylome-Wide Study and Potential Implications for Therapy. Int J Mol Sci 2024; 25:8094. [PMID: 39125664 PMCID: PMC11311678 DOI: 10.3390/ijms25158094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Kirsten Rat Sarcoma (KRAS) is the most commonly mutated oncogene in colorectal carcinoma (CRC). We have previously reported the interactions between microsatellite instability (MSI), DNA promoter methylation, and gene expression. In this study, we looked for associations between KRAS mutation, gene expression, and methylation that may help with precision medicine. Genome-wide gene expression and DNA methylation were done in paired CRC tumor and surrounding healthy tissues. The results suggested that (a) the magnitude of dysregulation of many major gene pathways in CRC was significantly greater in patients with the KRAS mutation, (b) the up- and down-regulation of these dysregulated gene pathways could be correlated with the corresponding hypo- and hyper-methylation, and (c) the up-regulation of CDKN2A was more pronounced in tumors with the KRAS mutation. A recent cell line study showed that there were higher CDKN2A levels in 5-FU-resistant CRC cells and that these could be down-regulated by Villosol. Our findings suggest the possibility of a better response to anti-CDKN2A therapy with Villosol in KRAS-mutant CRC. Also, the more marked up-regulation of genes in the proteasome pathway in CRC tissue, especially with the KRAS mutation and MSI, may suggest a potential role of a proteasome inhibitor (bortezomib, carfilzomib, or ixazomib) in selected CRC patients if necessary.
Collapse
Affiliation(s)
- Farzana Jasmine
- Institute for Population and Precision Health, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA; (A.A.); (Y.K.); (H.A.); (M.G.K.)
| | - Armando Almazan
- Institute for Population and Precision Health, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA; (A.A.); (Y.K.); (H.A.); (M.G.K.)
| | - Yuliia Khamkevych
- Institute for Population and Precision Health, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA; (A.A.); (Y.K.); (H.A.); (M.G.K.)
| | - Marc Bissonnette
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Habibul Ahsan
- Institute for Population and Precision Health, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA; (A.A.); (Y.K.); (H.A.); (M.G.K.)
- Department of Public Health Sciences, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Muhammad G. Kibriya
- Institute for Population and Precision Health, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA; (A.A.); (Y.K.); (H.A.); (M.G.K.)
- Department of Public Health Sciences, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
31
|
Liu HX, Feng J, Jiang JJ, Shen WJ, Zheng Y, Liu G, Gao XY. Integrated single-cell and bulk RNA sequencing revealed an epigenetic signature predicts prognosis and tumor microenvironment colorectal cancer heterogeneity. World J Gastrointest Oncol 2024; 16:3032-3054. [PMID: 39072180 PMCID: PMC11271797 DOI: 10.4251/wjgo.v16.i7.3032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/07/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) prognosis prediction is currently a major challenge. Epigenetic regulation has been widely reported for its role in cancer development. AIM To construct a robust prognostic signature, we used developed and validated across datasets. METHODS After constructing the signature, the prognostic value of the signature was evaluated in the TCGA cohort and six independent datasets (GSE17526, GSE17537, GSE33113, GSE37892, GSE39048 and GSE39582). The clinical, genomic and transcriptomic features related to the signature were identified. The correlations of the signature score with immune cell infiltration and cell-cell interactions were analyzed. The correlations between the signature score and the sensitivity to different drugs were also predicted. RESULTS In the TCGA cohort, patients in the low-risk group according to the signature score had longer survival than those in the high-risk group, and this finding was validated in the validation datasets. The signature was a prognostic factor independent of age and sex and was correlated with stage and PD-1/PD-L1 expression. Area under the receiving operating characteristic curve was 0.72. Genomic association analyses revealed that samples from high-risk patients exhibited chromosomal instability. Transcriptomic analyses revealed that the signature score was significantly associated with multiple cellular pathways. Bulk RNA-seq and single-cell sequencing data revealed that the signature reflected differences in infiltrating immune cell-tumor cell interactions, especially for macrophages. The signature also predicted the putative drug sensitivity of CRC samples. CONCLUSION The signature is a valuable biomarker for predicting CRC prognosis and reflects multiple features of CRC, especially macrophage infiltration in the microenvironment.
Collapse
Affiliation(s)
- Han-Xuan Liu
- Beijing Jinghua Anliang Technology, Beijing 102627, China
| | - Jie Feng
- Department of Clinical Laboratory, The First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jing-Jing Jiang
- Clinical Biological Sample Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Wan-Jun Shen
- Department of Nephrology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Zheng
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Gang Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiang-Yang Gao
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
32
|
Fialková V, Ďúranová H, Borotová P, Klongová L, Grabacka M, Speváková I. Natural Stilbenes: Their Role in Colorectal Cancer Prevention, DNA Methylation, and Therapy. Nutr Cancer 2024; 76:760-788. [PMID: 38950568 DOI: 10.1080/01635581.2024.2364391] [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: 03/05/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024]
Abstract
The resistance of colorectal cancer (CRC) to conventional therapeutic modalities, such as radiation therapy and chemotherapy, along with the associated side effects, significantly limits effective anticancer strategies. Numerous epigenetic investigations have unveiled that naturally occurring stilbenes can modify or reverse abnormal epigenetic alterations, particularly aberrant DNA methylation status, offering potential avenues for preventing or treating CRC. By modulating the activity of the DNA methylation machinery components, phytochemicals may influence the various stages of CRC carcinogenesis through multiple molecular mechanisms. Several epigenetic studies, especially preclinical research, have highlighted the effective DNA methylation modulatory effects of stilbenes with minimal adverse effects on organisms, particularly in combination therapies for CRC. However, the available preclinical and clinical data regarding the effects of commonly encountered stilbenes against CRC are currently limited. Therefore, additional epigenetic research is warranted to explore the preventive potential of these phytochemicals in CRC development and to validate their therapeutic application in the prevention and treatment of CRC. This review aims to provide an overview of selected bioactive stilbenes as potential chemopreventive agents for CRC with a focus on their modulatory mechanisms of action, especially in targeting alterations in DNA methylation machinery in CRC.
Collapse
Affiliation(s)
- Veronika Fialková
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Hana Ďúranová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Petra Borotová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Lucia Klongová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, Cracow, Poland
| | - Ivana Speváková
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| |
Collapse
|
33
|
Liang X, Aouizerat BE, So‐Armah K, Cohen MH, Marconi VC, Xu K, Justice AC. DNA methylation-based telomere length is associated with HIV infection, physical frailty, cancer, and all-cause mortality. Aging Cell 2024; 23:e14174. [PMID: 38629454 PMCID: PMC11258465 DOI: 10.1111/acel.14174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 07/21/2024] Open
Abstract
Telomere length (TL) is an important indicator of cellular aging. Shorter TL is associated with several age-related diseases including coronary heart disease, heart failure, diabetes, osteoporosis, and cancer. Recently, a DNA methylation-based TL (DNAmTL) estimator has been developed as an alternative method for directly measuring TL. In this study, we examined the association of DNAmTL with cancer prevalence and mortality risk among people with and without HIV in the Veterans Aging Cohort Study Biomarker Cohort (VACS, N = 1917) and Women's Interagency HIV Study Cohort (WIHS, N = 481). We profiled DNAm in whole blood (VACS) or in peripheral blood mononuclear cells (WIHS) using an array-based method. Cancer prevalence was estimated from electronic medical records and cancer registry data. The VACS Index was used as a measure of physiologic frailty. Models were adjusted for self-reported race and ethnicity, batch, smoking status, alcohol consumption, and five cell types (CD4, CD8, NK, B cell, and monocyte). We found that people with HIV had shorter average DNAmTL than those without HIV infection [beta = -0.25, 95% confidence interval (-0.32, -0.18), p = 1.48E-12]. Greater value of VACS Index [beta = -0.002 (-0.003, -0.001), p = 2.82E-05] and higher cancer prevalence [beta = -0.07 (-0.10, -0.03), p = 1.37E-04 without adjusting age] were associated with shortened DNAmTL. In addition, one kilobase decrease in DNAmTL was associated with a 40% increase in mortality risk [hazard ratio: 0.60 (0.44, 0.82), p = 1.42E-03]. In summary, HIV infection, physiologic frailty, and cancer are associated with shortening DNAmTL, contributing to an increased risk of all-cause mortality.
Collapse
Affiliation(s)
- Xiaoyu Liang
- Department of Epidemiology and BiostatisticsMichigan State UniversityEast LansingMichiganUSA
| | - Bradley E. Aouizerat
- Translational Research Center, College of DentistryNew York UniversityNew YorkNew YorkUSA
- Department of Oral and Maxillofacial Surgery, College of DentistryNew York UniversityNew YorkNew YorkUSA
| | - Kaku So‐Armah
- Boston University School of MedicineBostonMassachusettsUSA
| | - Mardge H. Cohen
- Department of MedicineStroger Hospital of Cook CountyChicagoIllinoisUSA
| | - Vincent C. Marconi
- Emory University School of Medicine and Rollins School of Public HealthThe Atlanta Veterans Affairs Medical CenterAtlantaGeorgiaUSA
| | - Ke Xu
- Department of PsychiatryYale School of MedicineNew HavenConnecticutUSA
- VA Connecticut Healthcare SystemWest HavenConnecticutUSA
| | - Amy C. Justice
- VA Connecticut Healthcare SystemWest HavenConnecticutUSA
- Department of Internal MedicineYale School of MedicineNew HavenConnecticutUSA
- Yale School of Public HealthNew HavenConnecticutUSA
| |
Collapse
|
34
|
Kafka A, Pećina-Šlaus N, Drmić D, Bukovac A, Njirić N, Žarković K, Jakovčević A. SFRP4 protein expression is reduced in high grade astrocytomas which is not caused by the methylation of its promoter. Front Mol Neurosci 2024; 17:1398872. [PMID: 38993819 PMCID: PMC11236799 DOI: 10.3389/fnmol.2024.1398872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction Epigenetics play a vital role in stratifying CNS tumors and gliomas. The importance of studying Secreted frizzled-related protein 4 (SFRP4) in gliomas is to improve diffuse glioma methylation profiling. Here we examined the methylation status of SFRP4 promoter and the level of its protein expression in diffuse gliomas WHO grades 2-4. Methods SFRP4 expression was detected by immunohistochemistry and evaluated semi-quantitatively. In the tumor hot-spot area, the intensity of protein expression in 200 cells was determined using ImageJ (National Institutes of Health, United States). The assessment of immunopositivity was based on the IRS score (Immunoreactivity Score). Promoter methylation was examined by methylation specific-PCR (MSP) in fifty-one diffuse glioma samples and appropriate controls. Isolated DNA was treated with bisulfite conversion and afterwards used for MSP. Public databases (cBioPortal, COSMIC and LOVD) were searched to corroborate the results. Results and discussion SFRP4 protein expression in glioblastomas was very weak or non-existent in 86.7% of samples, moderate in 13.3%, while strong expression was not observed. The increase in astrocytoma grade resulted in SFRP4 protein decrease (p = 0.008), indicating the loss of its antagonistic role in Wnt signaling. Promoter methylation of SFRP4 gene was found in 16.3% of cases. Astrocytomas grade 2 had significantly more methylated cases compared to grade 3 astrocytomas (p = 0.004) and glioblastomas (p < 0.001), which may indicate temporal niche of methylation in grade 2. Furthermore, the expression levels of SFRP4 were high in samples with methylated SFRP4 promoter and low or missing in unmethylated cases (Pearson's R = -0.413; p = 0.003). We also investigated the association of SFRP4 changes to key Wnt regulators GSK3β and DKK3 and established a positive correlation between methylations of SFRP4 and GSK3β (Pearson's R = 0.323; p = 0.03). Furthermore, SFRP4 expression was correlated to unmethylated DKK3 (Chi square = 7.254; p = 0.027) indication that Wnt signaling antagonist is associated to negative regulator's demethylation. Conclusion The study contributes to the recognition of the significance of epigenetic changes in diffuse glioma indicating that restoring SFRP4 protein holds potential as therapeutic avenue. Reduced expression of SFRP4 in glioblastomas, not following promoter methylation pattern, suggests another mechanism, possible global methylation, that turns off SFRP4 expression in higher grades.
Collapse
Affiliation(s)
- Anja Kafka
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nives Pećina-Šlaus
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Denis Drmić
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anja Bukovac
- Laboratory of Neuro-oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Niko Njirić
- Department of Neurosurgery, University Hospital Center “Zagreb”, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Kamelija Žarković
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Pathology, University Hospital Center “Zagreb”, Zagreb, Croatia
| | - Antonia Jakovčević
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Division of Pathology, University Hospital Center “Zagreb”, Zagreb, Croatia
| |
Collapse
|
35
|
Yu Z, Li J, Deng Y, Li C, Ye M, Zhang Y, Huang Y, Wang X, Zhao X, Liu J, Liu Z, Yin X, Mei L, Hou Y, Hu Q, Huang Y, Wang R, Fu H, Qiu R, Xu J, Gong Z, Zhang D, Zhang X. Lung tumor discrimination by deep neural network model CanDo via DNA methylation in bronchial lavage. iScience 2024; 27:110079. [PMID: 38883836 PMCID: PMC11176796 DOI: 10.1016/j.isci.2024.110079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/02/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Bronchoscopic-assisted discrimination of lung tumors presents challenges, especially in cases with contraindications or inaccessible lesions. Through meta-analysis and validation using the HumanMethylation450 database, this study identified methylation markers for molecular discrimination in lung tumors and designed a sequencing panel. DNA samples from 118 bronchial washing fluid (BWF) specimens underwent enrichment via multiplex PCR before targeted methylation sequencing. The Recursive Feature Elimination Cross-Validation and deep neural network algorithm established the CanDo classification model, which incorporated 11 methylation features (including 8 specific to the TBR1 gene), demonstrating a sensitivity of 98.6% and specificity of 97.8%. In contrast, bronchoscopic rapid on-site evaluation (bronchoscopic-ROSE) had lower sensitivity (87.7%) and specificity (80%). Further validation in 33 individuals confirmed CanDo's discriminatory potential, particularly in challenging cases for bronchoscopic-ROSE due to pathological complexity. CanDo serves as a valuable complement to bronchoscopy for the discriminatory diagnosis and stratified management of lung tumors utilizing BWF specimens.
Collapse
Affiliation(s)
- Zezhong Yu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jieyi Li
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing 314033, China
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Yi Deng
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chun Li
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Maosong Ye
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Zhang
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuqing Huang
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Xintao Wang
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing 314033, China
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Xiaokai Zhao
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing 314033, China
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Jie Liu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zilong Liu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xia Yin
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lijiang Mei
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingyong Hou
- Department of Pathology Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qin Hu
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yao Huang
- Department of Pulmonary, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210046, China
| | - Rongping Wang
- Department of Pulmonary, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210046, China
| | - Huiyu Fu
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Rumeng Qiu
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Jiahuan Xu
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
| | - Ziying Gong
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing 314033, China
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
- Department of R&D, Shanghai Yunying Biopharmaceutical Technology Co., Ltd., Shanghai 201612, China
| | - Daoyun Zhang
- Jiaxing Key Laboratory of Precision Medicine and Companion Diagnostics, Jiaxing Yunying Medical Inspection Co., Ltd., Jiaxing 314033, China
- Department of R&D, Zhejiang Yunying Medical Technology Co., Ltd., Jiaxing 314006, China
- Department of R&D, Shanghai Yunying Biopharmaceutical Technology Co., Ltd., Shanghai 201612, China
| | - Xin Zhang
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Pulmonary, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210046, China
| |
Collapse
|
36
|
Wu J, Li W, Su J, Zheng J, Liang Y, Lin J, Xu B, Liu Y. Integration of single-cell sequencing and bulk RNA-seq to identify and develop a prognostic signature related to colorectal cancer stem cells. Sci Rep 2024; 14:12270. [PMID: 38806611 PMCID: PMC11133358 DOI: 10.1038/s41598-024-62913-3] [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: 01/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
The prognosis for patients with colorectal cancer (CRC) remains worse than expected due to metastasis, recurrence, and resistance to chemotherapy. Colorectal cancer stem cells (CRCSCs) play a vital role in tumor metastasis, recurrence, and chemotherapy resistance. However, there are currently no prognostic markers based on CRCSCs-related genes available for clinical use. In this study, single-cell transcriptome sequencing was employed to distinguish cancer stem cells (CSCs) in the CRC microenvironment and analyze their properties at the single-cell level. Subsequently, data from TCGA and GEO databases were utilized to develop a prognostic risk model for CRCSCs-related genes and validate its diagnostic performance. Additionally, functional enrichment, immune response, and chemotherapeutic drug sensitivity of the relevant genes in the risk model were investigated. Lastly, the key gene RPS17 in the risk model was identified as a potential prognostic marker and therapeutic target for further comprehensive studies. Our findings provide new insights into the prognostic treatment of CRC and offer novel perspectives for a systematic and comprehensive understanding of CRC development.
Collapse
Affiliation(s)
- Jiale Wu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Wanyu Li
- Well Lead Medical Co., Ltd., Guangzhou, 511434, Guangdong, China
| | - Junyu Su
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiamin Zheng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Yanwen Liang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Jiansuo Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Bilian Xu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| | - Yi Liu
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
37
|
Zhou Y, Wang Q, Yin T, Zhao D, Zhou G, Sun X, Tan C, Zhou L, Yao S. Association Between Vitamin B12 Intake and Mortality in Patients with Colorectal Cancer: The US National Health and Nutrition Examination Survey, 1999-2018. Nutr Cancer 2024; 76:619-627. [PMID: 38775076 DOI: 10.1080/01635581.2024.2353938] [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: 12/11/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 07/02/2024]
Abstract
Vitamin B12 plays a role in DNA methylation, influencing the 1-carbon cycle; However, its effect on colorectal cancer (CRC) mortality remains uncertain. This study assessed the relationship between vitamin B12 intake and all-cause and cancer-specific mortality among CRC patients. We analyzed data from the NHANES from 1999 to 2018, using multivariable Cox regression, competing risk model, Kaplan-Meier survival curves, and stratified analysis with interaction effects. The studied involved 4,554 cancer patients (mean age 65.8 years, 47.6% males). Results from multivariate Cox regression indicated that each additional 1 mcg/day of dietary vitamin B12 independently increased the risk of all-cause (HR, 1.07; 95% CI: 1.04-1.09, p < 0.001) and cancer-specific mortality (HR, 1.04; 95% CI, 1.02-1.06; p < 0.001). Kaplan-Meier curves indicated a higher risk of all-cause mortality with increased vitamin B12 intake (Log rank p = 0.01). Subgroup analysis suggested that higher vitamin B12 intake correlated with increased all-cause mortality risk, especially in individuals with higher protein (HR, 1.04; 95% CI, 1.02-1.06; p = 0.019) or carbohydrate intake (HR, 1.03; 95% CI, 1.01-1.05; p = 0.04). Thus, higher vitamin B12 intake correlates with increased all-cause and cancer-specific mortality in CRC patients, particularly those with higher protein or carbohydrate intake.
Collapse
Affiliation(s)
- Yuanchen Zhou
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Qianqian Wang
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Tengfei Yin
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Dongyan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Geyujia Zhou
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xizhen Sun
- Department of Gastroenterology, Beijing Jishuitan Hospital, Beijing, China
| | - Chang Tan
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Lei Zhou
- Department of Gastrointestinal Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Shukun Yao
- Graduate School, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
38
|
Parreno V, Loubiere V, Schuettengruber B, Fritsch L, Rawal CC, Erokhin M, Győrffy B, Normanno D, Di Stefano M, Moreaux J, Butova NL, Chiolo I, Chetverina D, Martinez AM, Cavalli G. Transient loss of Polycomb components induces an epigenetic cancer fate. Nature 2024; 629:688-696. [PMID: 38658752 PMCID: PMC11096130 DOI: 10.1038/s41586-024-07328-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2024] [Indexed: 04/26/2024]
Abstract
Although cancer initiation and progression are generally associated with the accumulation of somatic mutations1,2, substantial epigenomic alterations underlie many aspects of tumorigenesis and cancer susceptibility3-6, suggesting that genetic mechanisms might not be the only drivers of malignant transformation7. However, whether purely non-genetic mechanisms are sufficient to initiate tumorigenesis irrespective of mutations has been unknown. Here, we show that a transient perturbation of transcriptional silencing mediated by Polycomb group proteins is sufficient to induce an irreversible switch to a cancer cell fate in Drosophila. This is linked to the irreversible derepression of genes that can drive tumorigenesis, including members of the JAK-STAT signalling pathway and zfh1, the fly homologue of the ZEB1 oncogene, whose aberrant activation is required for Polycomb perturbation-induced tumorigenesis. These data show that a reversible depletion of Polycomb proteins can induce cancer in the absence of driver mutations, suggesting that tumours can emerge through epigenetic dysregulation leading to inheritance of altered cell fates.
Collapse
Affiliation(s)
- V Parreno
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - V Loubiere
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - B Schuettengruber
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - L Fritsch
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - C C Rawal
- University of Southern California, Los Angeles, CA, USA
| | - M Erokhin
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - B Győrffy
- Semmelweis University Department of Bioinformatics, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| | - D Normanno
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - M Di Stefano
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - J Moreaux
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- UFR Medicine, University of Montpellier, Montpellier, France
| | - N L Butova
- University of Southern California, Los Angeles, CA, USA
| | - I Chiolo
- University of Southern California, Los Angeles, CA, USA
| | - D Chetverina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A-M Martinez
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France.
| | - G Cavalli
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France.
| |
Collapse
|
39
|
Zhao N, Lai C, Wang Y, Dai S, Gu H. Understanding the role of DNA methylation in colorectal cancer: Mechanisms, detection, and clinical significance. Biochim Biophys Acta Rev Cancer 2024; 1879:189096. [PMID: 38499079 DOI: 10.1016/j.bbcan.2024.189096] [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: 10/05/2023] [Revised: 02/18/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Colorectal cancer (CRC) is one of the deadliest malignancies worldwide, ranking third in incidence and second in mortality. Remarkably, early stage localized CRC has a 5-year survival rate of over 90%; in stark contrast, the corresponding 5-year survival rate for metastatic CRC (mCRC) is only 14%. Compounding this problem is the staggering lack of effective therapeutic strategies. Beyond genetic mutations, which have been identified as critical instigators of CRC initiation and progression, the importance of epigenetic modifications, particularly DNA methylation (DNAm), cannot be underestimated, given that DNAm can be used for diagnosis, treatment monitoring and prognostic evaluation. This review addresses the intricate mechanisms governing aberrant DNAm in CRC and its profound impact on critical oncogenic pathways. In addition, a comprehensive review of the various techniques used to detect DNAm alterations in CRC is provided, along with an exploration of the clinical utility of cancer-specific DNAm alterations.
Collapse
Affiliation(s)
- Ningning Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Chuanxi Lai
- Division of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Yunfei Wang
- Zhejiang ShengTing Biotech. Ltd, Hangzhou 310000, China
| | - Sheng Dai
- Division of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China.
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China.
| |
Collapse
|
40
|
Gao J, Shi W, Wang J, Guan C, Dong Q, Sheng J, Zou X, Xu Z, Ge Y, Yang C, Li J, Bao H, Zhong X, Cui Y. Research progress and applications of epigenetic biomarkers in cancer. Front Pharmacol 2024; 15:1308309. [PMID: 38681199 PMCID: PMC11048075 DOI: 10.3389/fphar.2024.1308309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
Epigenetic changes are heritable changes in gene expression without changes in the nucleotide sequence of genes. Epigenetic changes play an important role in the development of cancer and in the process of malignancy metastasis. Previous studies have shown that abnormal epigenetic changes can be used as biomarkers for disease status and disease prediction. The reversibility and controllability of epigenetic modification changes also provide new strategies for early disease prevention and treatment. In addition, corresponding drug development has also reached the clinical stage. In this paper, we will discuss the recent progress and application status of tumor epigenetic biomarkers from three perspectives: DNA methylation, non-coding RNA, and histone modification, in order to provide new opportunities for additional tumor research and applications.
Collapse
Affiliation(s)
- Jianjun Gao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wujiang Shi
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiangang Wang
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Canghai Guan
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jialin Sheng
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinlei Zou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoqiang Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengru Yang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiehan Li
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Ma Y, Li J, Zhao X, Ji C, Hu W, Ma Y, Qu F, Sun Y, Zhang X. Multi-omics cluster defines the subtypes of CRC with distinct prognosis and tumor microenvironment. Eur J Med Res 2024; 29:207. [PMID: 38549156 PMCID: PMC10976740 DOI: 10.1186/s40001-024-01805-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a complex malignancy characterized by diverse molecular profiles, clinical outcomes, and limited precision in prognostic markers. Addressing these challenges, this study utilized multi-omics data to define consensus molecular subtypes in CRC and elucidate their association with clinical outcomes and underlying biological processes. METHODS Consensus molecular subtypes were obtained by applying ten integrated multi-omics clustering algorithms to analyze TCGA-CRC multi-omics data, including mRNA, lncRNA, miRNA, DNA methylation CpG sites, and somatic mutation data. The association of subtypes with prognoses, enrichment functions, immune status, and genomic alterations were further analyzed. Next, we conducted univariate Cox and Lasso regression analyses to investigate the potential prognostic application of biomarkers associated with multi-omics subtypes derived from weighted gene co-expression network analysis (WGCNA). The function of one of the biomarkers MID2 was validated in CRC cell lines. RESULTS Two CRC subtypes linked to distinct clinical outcomes were identified in TCGA-CRC cohort and validated with three external datasets. The CS1 subtype exhibited a poor prognosis and was characterized by higher tumor-related Hallmark pathway activity and lower metabolism pathway activity. In addition, the CS1 was predicted to have less immunotherapy responder and exhibited more genomic alteration compared to CS2. Then a prognostic model comprising five genes was established, with patients in the high-risk group showing substantial concordance with the CS1 subtype, and those in the low-risk group with the CS2 subtype. The gene MID2, included in the prognostic model, was found to be correlated with epithelial-mesenchymal transition (EMT) pathway and distinct DNA methylation patterns. Knockdown of MID2 in CRC cells resulted in reduced colony formation, migration, and invasion capacities. CONCLUSION The integrative multi-omics subtypes proposed potential biomarkers for CRC and provided valuable knowledge for precision oncology.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Jing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Xu Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Chao Ji
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Weibin Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - YanFang Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Fengyi Qu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Yuchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China
| | - Xiaozhi Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Yanta West Road 277, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
42
|
Huang H, Li Q, Tu X, Yu D, Zhou Y, Ma L, Wei K, Gao Y, Zhao G, Han R, Ye F, Ke C. DNA hypomethylation patterns and their impact on the tumor microenvironment in colorectal cancer. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00933-x. [PMID: 38520647 DOI: 10.1007/s13402-024-00933-x] [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] [Accepted: 03/02/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Recent research underscores the pivotal role of immune checkpoints as biomarkers in colorectal cancer (CRC) therapy, highlighting the dynamics of resistance and response to immune checkpoint inhibitors. The impact of epigenetic alterations in CRC, particularly in relation to immune therapy resistance, is not fully understood. METHODS We integrated a comprehensive dataset encompassing TCGA-COAD, TCGA-READ, and multiple GEO series (GSE14333, GSE37892, GSE41258), along with key epigenetic datasets (TCGA-COAD, TCGA-READ, GSE77718). Hierarchical clustering, based on Euclidean distance and Ward's method, was applied to 330 primary tumor samples to identify distinct clusters. The immune microenvironment was assessed using MCPcounter. Machine learning algorithms were employed to predict DNA methylation patterns and their functional enrichment, in addition to transcriptome expression analysis. Genomic mutation profiles and treatment response assessments were also conducted. RESULTS Our analysis delineated a specific tumor cluster with CpG Island (CGI) methylation, termed the Demethylated Phenotype (DMP). DMP was associated with metabolic pathways such as oxidative phosphorylation, implicating increased ATP production efficiency in mitochondria, which contributes to tumor aggressiveness. Furthermore, DMP showed activation of the Myc target pathway, known for tumor immune suppression, and exhibited downregulation in key immune-related pathways, suggesting a tumor microenvironment characterized by diminished immunity and increased fibroblast infiltration. Six potential therapeutic agents-lapatinib, RDEA119, WH.4.023, MG.132, PD.0325901, and AZ628-were identified as effective for the DMP subtype. CONCLUSION This study unveils a novel epigenetic phenotype in CRC linked to resistance against immune checkpoint inhibitors, presenting a significant step toward personalized medicine by suggesting epigenetic classifications as a means to identify ideal candidates for immunotherapy in CRC. Our findings also highlight potential therapeutic agents for the DMP subtype, offering new avenues for tailored CRC treatment strategies.
Collapse
Grants
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
Collapse
Affiliation(s)
- He Huang
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Qian Li
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xusheng Tu
- Department of Emergency Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Dongyue Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yundong Zhou
- Shanghai Medical Innovation Fusion Biomedical Research Center, Shanghai, China
| | - Lifei Ma
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- College of Lab Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Kongyuan Wei
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Guodong Zhao
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China
| | - Ruiqin Han
- State Key Laboratory of Common Mechanism Research for Major Diseas, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200000, China.
| | - Chunlian Ke
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China.
| |
Collapse
|
43
|
Cho SH, Kim YM, An HJ, Kim JH, Kim NK. miR-665-Mediated Regulation of AHCYL2 and BVES Genes in Recurrent Implantation Failure. Genes (Basel) 2024; 15:244. [PMID: 38397233 PMCID: PMC10888078 DOI: 10.3390/genes15020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
The primary goal of this investigation was to identify mRNA targets affected by dysregulated miRNAs in RIF. This was accomplished by comprehensively analyzing mRNA and miRNA expression profiles in two groups: female subjects with normal reproductive function (control, n = 5) and female subjects experiencing recurrent implantation failure (RIF, n = 5). We conducted transcriptome sequencing and small RNA sequencing on endometrial tissue samples from these cohorts. Subsequently, we validated a selection of intriguing findings using real-time PCR with samples from the same cohort. In total, our analysis revealed that 929 mRNAs exhibited differential expression patterns between the control and RIF patient groups. Notably, our investigation confirmed the significant involvement of dysregulated genes in the context of RIF. Furthermore, we uncovered promising correlation patterns within these mRNA/miRNA pairs. Functional categorization of these miRNA/mRNA pairs highlighted that the differentially expressed genes were predominantly associated with processes such as angiogenesis and cell adhesion. We identified new target genes that are regulated by miR-665, including Blood Vessel Epicardial Substance (BVES) and Adenosylhomocysteinase like 2 (AHCYL2). Our findings suggest that abnormal regulation of genes involved in angiogenesis and cell adhesion, including BVES and AHCYL2, contributes to the endometrial dysfunction observed in women with recurrent implantation failure (RIF) compared to healthy women.
Collapse
Affiliation(s)
- Sung Hwan Cho
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (S.H.C.); (H.J.A.)
- College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Young Myeong Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Hui Jeong An
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (S.H.C.); (H.J.A.)
- College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam 13496, Republic of Korea
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea; (S.H.C.); (H.J.A.)
| |
Collapse
|
44
|
Shim H, Jang K, Bang YH, Chu HBK, Kang J, Lee JY, Cho S, Lee HS, Jeon J, Hwang T, Joe S, Lim J, Choi JH, Joo EH, Park K, Moon JH, Han KY, Hong Y, Lee WY, Kim HC, Yun SH, Cho YB, Park YA, Huh JW, Shin JK, Pyo DH, Hong H, Lee HO, Park WY, Yang JO, Kim YJ. Comprehensive profiling of DNA methylation in Korean patients with colorectal cancer. BMB Rep 2024; 57:110-115. [PMID: 37605617 PMCID: PMC10910091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/27/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023] Open
Abstract
Alterations in DNA methylation play an important pathophysiological role in the development and progression of colorectal cancer. We comprehensively profiled DNA methylation alterations in 165 Korean patients with colorectal cancer (CRC), and conducted an in-depth investigation of cancer-specific methylation patterns. Our analysis of the tumor samples revealed a significant presence of hypomethylated probes, primarily within the gene body regions; few hypermethylated sites were observed, which were mostly enriched in promoter-like and CpG island regions. The CpG Island Methylator PhenotypeHigh (CIMP-H) exhibited notable enrichment of microsatellite instability-high (MSI-H). Additionally, our findings indicated a significant correlation between methylation of the MLH1 gene and MSI-H status. Furthermore, we found that the CIMP-H had a higher tendency to affect the right-side of the colon tissues and was slightly more prevalent among older patients. Through our methylome profile analysis, we successfully verified the thylation patterns and clinical characteristics of Korean patients with CRC. This valuable dataset lays a strong foundation for exploring novel molecular insights and potential therapeutic targets for the treatment of CRC. [BMB Reports 2024; 57(2): 110-115].
Collapse
Affiliation(s)
- Hyeran Shim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Kiwon Jang
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Yeong Hak Bang
- Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hoang Bao Khanh Chu
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jisun Kang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jin-Young Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Sheehyun Cho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Hong Seok Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jongbum Jeon
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Taeyeon Hwang
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Soobok Joe
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Jinyeong Lim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06355, Korea
| | - Ji-Hye Choi
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06355, Korea
| | - Eun Hye Joo
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06355, Korea
| | - Kyunghee Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, Seoul 04779, Korea
| | - Ji Hwan Moon
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, Seoul 04779, Korea
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, Seoul 04779, Korea
| | - Yourae Hong
- Department of Oncology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium, Seoul 04779, Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Seong Hyeon Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yoon Ah Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jung Wook Huh
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jung Kyong Shin
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Dae Hee Pyo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hyekyung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hae-Ock Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Woong-Yang Park
- Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06355, Korea
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, Seoul 04779, Korea
| | - Jin Ok Yang
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Young-Joon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
- LepiDyne Co., Ltd., Seoul 04779, Korea
| |
Collapse
|
45
|
Ye J, Zhang J, Ding W. DNA methylation modulates epigenetic regulation in colorectal cancer diagnosis, prognosis and precision medicine. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:34-53. [PMID: 38464391 PMCID: PMC10918240 DOI: 10.37349/etat.2024.00203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/11/2023] [Indexed: 03/12/2024] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease influenced by the interplay of genetic and environmental factors. The clinical heterogeneity of CRC cannot be attributed exclusively to genetic diversity and environmental exposures, and epigenetic markers, especially DNA methylation, play a critical role as key molecular markers of cancer. This review compiles a comprehensive body of evidence underscoring the significant involvement of DNA methylation modifications in the pathogenesis of CRC. Moreover, this review explores the potential utility of DNA methylation in cancer diagnosis, prognostics, assessment of disease activity, and prediction of drug responses. Recognizing the impact of DNA methylation will enhance the ability to identify distinct CRC subtypes, paving the way for personalized treatment strategies and advancing precision medicine in the management of CRC.
Collapse
Affiliation(s)
- Jingxin Ye
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
- Department of Gastroenterology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian 223800, Jiangsu Province, China
| | - Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Weifeng Ding
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
46
|
Sacdalan DB, Ul Haq S, Lok BH. Plasma Cell-Free Tumor Methylome as a Biomarker in Solid Tumors: Biology and Applications. Curr Oncol 2024; 31:482-500. [PMID: 38248118 PMCID: PMC10814449 DOI: 10.3390/curroncol31010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/30/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
DNA methylation is a fundamental mechanism of epigenetic control in cells and its dysregulation is strongly implicated in cancer development. Cancers possess an extensively hypomethylated genome with focal regions of hypermethylation at CPG islands. Due to the highly conserved nature of cancer-specific methylation, its detection in cell-free DNA in plasma using liquid biopsies constitutes an area of interest in biomarker research. The advent of next-generation sequencing and newer computational technologies have allowed for the development of diagnostic and prognostic biomarkers that utilize methylation profiling to diagnose disease and stratify risk. Methylome-based predictive biomarkers can determine the response to anti-cancer therapy. An additional emerging application of these biomarkers is in minimal residual disease monitoring. Several key challenges need to be addressed before cfDNA-based methylation biomarkers become fully integrated into practice. The first relates to the biology and stability of cfDNA. The second concerns the clinical validity and generalizability of methylation-based assays, many of which are cancer type-specific. The third involves their practicability, which is a stumbling block for translating technologies from bench to clinic. Future work on developing pan-cancer assays with their respective validities confirmed using well-designed, prospective clinical trials is crucial in pushing for the greater use of these tools in oncology.
Collapse
Affiliation(s)
- Danielle Benedict Sacdalan
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 2374, Toronto, ON M5S 1A8, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2C4, Canada
| | - Sami Ul Haq
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2C4, Canada
- Schulich School of Medicine & Dentistry, Western University, 1151 Richmond St, London, ON N6A 5C1, Canada
| | - Benjamin H. Lok
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, 1 King’s College Circle, Medical Sciences Building, Room 2374, Toronto, ON M5S 1A8, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Room 15-701, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
47
|
Zhang K, He Q, Cao Q, Chuan J, Qin A, Tang L, Zhang X, Xiao C, Zhu B, Hu M, Chang L, Bu ZX, Fu L, Yang T, Wang Y, Liu W. Evaluating the clinical performance of SDC2/NDRG4 methylation for colorectal cancer detection. Epigenomics 2024; 16:93-108. [PMID: 38226561 DOI: 10.2217/epi-2023-0290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
Purpose: The performance and clinical accuracy of combined SDC2/NDRG4 methylation were evaluated in diagnosing colorectal cancer (CRC) and advanced adenoma. Methods: A total of 2333 participants were enrolled to assess the sensitivity and specificity of biomarkers in diagnosing CRC in a multicenter clinical trial through feces DNA methylation tests. Results: SDC2/NDRG4 methylation showed excellent performance for CRC detection in biomarker research and the real world. Its sensitivity for detecting CRC, early CRC and advanced adenoma were 92.06%, 91.45% and 62.61%, respectively. Its specificity was 94.29%, with a total coincidence rate of 88.28%. When interference samples were included, the specificity was still good (82.61%). Therefore, the SDC2/NDRG4 methylation test showed excellent performance in detecting CRC and advanced adenoma under clinical application.
Collapse
Affiliation(s)
- Ke Zhang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410000, China
| | - Qing He
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Qin Cao
- Department of Gastroenterology, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Jun Chuan
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Ang Qin
- Department of Endoscope Center, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Lin Tang
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Xinyue Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Changhe Xiao
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Biyin Zhu
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Meiling Hu
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Lei Chang
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Zhong Xin Bu
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Lanqi Fu
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Ting Yang
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
| | - Yu Wang
- GeneTalks Biotech Co., Ltd, Changsha, Hunan, 410000, PR China
- School of Life Sciences & Technology, Tongji University, Shanghai, 200092, China
| | - Weidong Liu
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410000, China
| |
Collapse
|
48
|
Sarangi J, Das P, Ahmad A, Sulaiman M, Ghosh S, Gupta B, Panwar R, Pal S, Yadav R, Ahuja V, Sen S, Upadhyay AD, Dash NR, Sharma A, Gupta SD. Methylation study of tumor suppressor genes in human aberrant crypt foci, colorectal carcinomas, and normal colon. J Cancer Res Ther 2024; 20:268-274. [PMID: 38554332 DOI: 10.4103/jcrt.jcrt_1573_22] [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: 07/28/2022] [Accepted: 09/22/2022] [Indexed: 04/01/2024]
Abstract
BACKGROUND Aberrant crypt foci (ACF) are the earliest preneoplastic lesions in human colon, identifiable on chromoendoscopic screening. Our objective was to evaluate the %methylation of APC, CDKN2A, MLH1, RASSF1, MGMT, and WIF1 tumor suppressor genes (TSG) in ACF, corresponding colorectal carcinomas (CRC), and normal colonic mucosal controls. METHODS In this study, macroscopically normal-appearing mucosal flaps were sampled 5-10 cm away from the tumor mass from 302 fresh colectomy specimens to identify ACF-like lesions. Thirty-five cases with multiple ACFs were selected (n 35) as the main study group, with corresponding sections from CRC (n 35) as disease controls, and mucosal tissue blocks from 20 colectomy specimens (normal controls), operated for non-neoplastic pathologies. Genomic DNA was extracted, and methylation-specific polymerase chain reaction (PCR) was performed on a customized methylation array model. %Methylation data were compared among the groups and with clinicopathological parameters. Selected target mRNA and protein expression studies were performed. RESULTS %Methylation of TSGs in ACF was intermediate between normal colon and CRC, although a statistically significant difference was observed only for the WIF1 gene (P < 0.01). Also, there was increased nuclear β-catenin expression and upregulation of CD44-positive cancer-stem cells in ACF and CRCs than in controls. Right-sided ACFs and dysplastic ACFs had a higher %methylation of CDKN2A (P < 0.01), whereas hyperplastic ACFs had a higher %methylation of RASSF1 (P 0.04). The topographic characteristics of ACFs did not correlate with TSG %methylation. CONCLUSIONS Early epigenetic methylation of WIF1 gene is one of the mechanisms for ACF development in human colon.
Collapse
Affiliation(s)
- Jayati Sarangi
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Aijaz Ahmad
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohamed Sulaiman
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Shouriyo Ghosh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Brijnandan Gupta
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Panwar
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Sujoy Pal
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vineet Ahuja
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Sudip Sen
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asish D Upadhyay
- Department of Biostatistics and, All India Institute of Medical Sciences, New Delhi, India
| | - Nihar R Dash
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Sharma
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Siddhartha D Gupta
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
49
|
Huang S, Chang S, Liao T, Yang M. Detection and clinical significance of CEACAM5 methylation in colorectal cancer patients. Cancer Sci 2024; 115:270-282. [PMID: 37942534 PMCID: PMC10823287 DOI: 10.1111/cas.16012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
Colorectal cancer (CRC) is a globally common cancer, and the serum carcinoembryonic antigen (sCEA) is widely applied as a diagnostic and prognostic tumor marker in CRC. This study aimed to elucidate the mechanism of CEA expression and corresponding clinical features to improve prognostic assessments. In CRC cells, hypomethylation of the CEACAM5 promoter enhanced CEA expression in HCT116 and HT29 cells with 5-aza-2'-deoxycytidine (5-Aza-dC) treatment. Our clinical data indicated that 64.7% (101/156) of CRC patients had an sCEA level above the normal range, and 76.2% (77/101) of those patients showed a lower average CpG methylation level of the CEACAM5 promoter. The methylation analysis showed that both CRC cell lines and patient samples shared the same critical methylation CpG regions at -200 to -500 and -1000 to -1400 bp of the CEACAM5 promoter. Patients with hypermethylation of the CEACAM5 promoter showed features of a BRAF mutation, TGFB2 mutation, microsatellite instability-high, and preference for right-sided colorectal cancer and peritoneal seeding presentation that had a similar clinical character to the consensus molecular subtype 1 (CMS1) of colorectal cancer. Additionally, hypermethylation of the CEACAM5 promoter combined with evaluated sCEA demonstrated the worst survival among the patients. Therefore, the methylation status of the CEACAM5 promoter also served as an effective biomarker for assessing disease prognosis. Results indicated that DNA methylation is a major regulatory mechanism for CEA expression in colorectal cancer. Moreover, our data also highlighted that patients in a subgroup who escaped from inactivation by DNA methylation had distinct clinical and pathological features and the worst survival.
Collapse
Affiliation(s)
- Sheng‐Chieh Huang
- Institute of Clinical MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Colorectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
- Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Shih‐Ching Chang
- Division of Colorectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
- Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tsai‐Tsen Liao
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipeiTaiwan
- Cancer Research CenterTaipei Medical University HospitalTaipeiTaiwan
| | - Muh‐Hwa Yang
- Institute of Clinical MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Cancer and Immunology Research CenterNational Yang Ming Chiao UniversityTaipeiTaiwan
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| |
Collapse
|
50
|
Erfanian N, Nasseri S, Miraki Feriz A, Safarpour H, Namaei MH. Characterization of Wnt signaling pathway under treatment of Lactobacillus acidophilus postbiotic in colorectal cancer using an integrated in silico and in vitro analysis. Sci Rep 2023; 13:22988. [PMID: 38151510 PMCID: PMC10752892 DOI: 10.1038/s41598-023-50047-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023] Open
Abstract
Colorectal cancer (CRC) is a prevalent and life-threatening cancer closely associated with the gut microbiota. Probiotics, as a vital microbiota group, interact with the host's colonic epithelia and immune cells by releasing a diverse range of metabolites named postbiotics. The present study examined the effects of postbiotics on CRC's prominent differentially expressed genes (DEGs) using in silico and in vitro analysis. Through single-cell RNA sequencing (scRNA-seq), we identified four DEGs in CRC, including secreted frizzled-related protein 1 (SFRP1), secreted frizzled-related protein 2 (SFRP2), secreted frizzled-related protein 4 (SFRP4), and matrix metallopeptidase 7 (MMP7). Enrichment analysis and ExpiMap, a novel deep learning-based method, determined that these DEGs are involved in the Wnt signaling pathway as a primary cascade in CRC. Also, spatial transcriptome analysis showed specific expression patterns of the SFRP2 gene in fibroblast cell type. The expression of selected DEGs was confirmed on CRC and normal adjacent tissues using Real-Time quantitative PCR (RT-qPCR). Moreover, we examined the effects of postbiotics extracted from Lactobacillus acidophilus (L. acidophilus) on the proliferation, migration, and cell cycle distribution of HT-29 cells using MTT, scratch, and flow cytometry assays. Our results showed that L. acidophilus postbiotics induce cell cycle arrest at G1 phase and also had anti-proliferative and anti-migration effects on HT-29 cells, while it did not exert anti-proliferative activity on control fibroblasts. Finally, we revealed that treating HT-29 cells with postbiotics can affect the expression of selected DEGs. We suggested that L. acidophilus postbiotics have therapeutic potential in CRC by modulating key genes in the Wnt pathway.
Collapse
Affiliation(s)
- Nafiseh Erfanian
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Nasseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Adib Miraki Feriz
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Mohammad Hassan Namaei
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|